WO2001088141A2 - Synthetic gagpol genes and their uses - Google Patents

Synthetic gagpol genes and their uses Download PDF

Info

Publication number
WO2001088141A2
WO2001088141A2 PCT/EP2001/005744 EP0105744W WO0188141A2 WO 2001088141 A2 WO2001088141 A2 WO 2001088141A2 EP 0105744 W EP0105744 W EP 0105744W WO 0188141 A2 WO0188141 A2 WO 0188141A2
Authority
WO
WIPO (PCT)
Prior art keywords
encoded
gag
nucleic acid
gagpol
gene
Prior art date
Application number
PCT/EP2001/005744
Other languages
French (fr)
Other versions
WO2001088141A3 (en
Inventor
Ralf Wagner
Markus Graf
Ludwig Deml
Kurt Bieler
Original Assignee
Geneart Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=8168767&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2001088141(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CA002408433A priority Critical patent/CA2408433A1/en
Priority to AU2001281781A priority patent/AU2001281781A1/en
Priority to US10/276,482 priority patent/US7378515B2/en
Priority to BR0110706-2A priority patent/BR0110706A/en
Priority to EP01960229A priority patent/EP1282712B1/en
Application filed by Geneart Gmbh filed Critical Geneart Gmbh
Priority to DE60121572T priority patent/DE60121572T2/en
Publication of WO2001088141A2 publication Critical patent/WO2001088141A2/en
Publication of WO2001088141A3 publication Critical patent/WO2001088141A3/en
Priority to HK03105174A priority patent/HK1053853A1/en
Priority to US11/754,755 priority patent/US8287881B2/en
Priority to US13/615,587 priority patent/US20130122585A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • Retroviral based vector particles for gene therapy, retroviral based virus-like particles for vaccination or retroviral based DNA or RNA constructs for DNA vaccination should require the ability to express structural genes and enzymatic functions in high yields in order to obtain maximal efficiency and yet, at the same time, with lowest possible risk and side effects for the recipient.
  • Efficient production of structural or enzymatic proteins from complex retroviruses like HTLN-1 and -2, Lentiviruses and Spumaviruses is limited by their complex regulatory mechanism of transcription, nuclear R ⁇ A translocation and expression.
  • SIN and HIN-1 late gene products is highly regulated both on transcnptional and post-transcriptional level therefore depending on the presence and function of early phase proteins like Tat and Rev.
  • lentiviral Gag and GagPol based vector particles Another preferred application of lentiviral Gag and GagPol based vector particles is gene transfer into different types of dividing, resting or postmitotic cells.
  • lentiviral gene transfer can be used for gene delivery into a variety of quiescent or non-dividing cells, such as neuronal, muscle, and liver as well as hematopoietic stem cells and might therefore considerably contribute to the prevention and treatment of genetic disorders, tumor diseases and infections.
  • FIG. 10 is a diagrammatic representation of FIG. 10
  • transgene refers to a nucleic acid which should be introduced in a cell as a therapeutic agent.
  • This nucleic acid can serve e.g. as a decoy, triplehelix forming or antisense oligonucleotide, as a ribozyme, an aptamere, or as a nucleic acid inserted into the genom in a manner that ensures its function, replication and transmission as a normal gene.
  • the present invention is based on the observation, that the codon usage e.g. in the HIN genome (A/T content 55.88 %) significantly deviates from that of highly expressed mammalian genes (usually not exceeding 50%) indicating a regulatory function of the codon usage for timely regulated retroviral such as HTLN-1 and -2, onco viral, spumaviral and lentiviral gene expression.
  • GagPol expression may be either constitutive or inducible e.g. by addition of a specific inducer like ecdysone in the case of a hormone inducible promotor enhancer or by removal of a repressor protein (Tet on/off) or repressor gene (Cre/Lox).
  • a specific inducer like ecdysone in the case of a hormone inducible promotor enhancer or by removal of a repressor protein (Tet on/off) or repressor gene (Cre/Lox).
  • transduction competent vector particles as well as infectious, although not necessarily replication competent, bacterial or viral vehicle encoding the synGagPol derived packaging proteins, and any appropriate envelope or receptor structure as well as the desired transgene may be used for application ex vivo and in vivo.
  • the present invention relates to nucleic acid molecules comprising the nucleic acid sequence according to the invention for use as an active pharmaceutical substance.
  • the Gag or GagPol based synthetic gene is modified in such a way that the active site of the reverse transcriptase gene (RT) is dislocated e.g. at the very C-terminus of the synthetic GagPol gene in order to inhibit polymerase activity thereby preventing the production of potentially hazardous nucleotide sequences and/or reconstitution of infectious or by other means hazardous virus particles.
  • RT reverse transcriptase gene
  • the delivery of codon optimized GagPol genes or derivatives thereof into cells in vitro may be mediated by any means of transfecting or introducing a construct, e.g. a plasmid or plasmid derivative such as "midges” (closed linear D ⁇ A) encoding the synthetic GagPol gene or the above variants thereof into cells.
  • a construct e.g. a plasmid or plasmid derivative such as "midges" (closed linear D ⁇ A) encoding the synthetic GagPol gene or the above variants thereof into cells.
  • the delivery of codon optimized GagPol genes or derivatives into cells in vitro and in vivo may be also mediated by infectious or non infectious recombinant viral vehicles such as recombinant Retroviruses, Vaccinia- or Poxviruses, Adenoviruses, Alphaviruses, Herpesviruses, Baculoviruses or any other recombinant virus, subviral components bridging transfection and infection procedures like e.g. Virosomes comprising nucleic acid/protein aggregates containing e.g. Influenca hemagglutinin, or bacterial vectors such as recombinant Listeriae or Salmonellae or any other type of infectious, replicating or non-replicating vector.
  • infectious or non infectious recombinant viral vehicles such as recombinant Retroviruses, Vaccinia- or Poxviruses, Adenoviruses, Alphaviruses, Herpesviruses, Baculoviruses or any other recombinant
  • Serum was recovered from mice by tail bleed at week two after the boost injection.
  • Antibodies specific for the HIV Pr55 gag polyprotein were quantified by an end-point dilution ELISA assay (in duplicate) on samples from individual animals.
  • a solid phase of PrepCell purified Pr55 g g proteins (100 ⁇ l of 1 ⁇ g/ml per well, overnight at 4° C in a refrigerator) was used to capture Pr55 gag -reactive antibodies from the serum (2 h at 37°C), which were then detected with horseradish peroxidase (HRP)-conjugated goat anti-mouse IgGl, IgG2a and total IgG antibodies (1:2000 in PBS, 2% Tween 20, 3% FCS; 100 ⁇ l/well; PharMingen, Hamburg, Germany), followed by o-phenlyenediamine dihydrochloride solution (100 ⁇ l/well, 20 min at room temperature in the dark; Abbott Laboratories, Abbott Park, IL).
  • RCR replication competent recombinants
  • CEMxl74 cells which support replication of SIV and SIV hybrid viruses utilising a heterologous env gene (Reiprich et al, 1997, J.Virol. 71, 3328-3331). These cells were infected with 0.5 ml of NiG ⁇ BH vector preparations (for titers see example 6, Table 2) packaged by Sgp ⁇ 2, Sgp syn , or Hgp s n and pHIT-G. Slightly elevated capsid antigen levels ( ⁇ 400 pg SINp27CA/ml) could be detected one and four days after infection.
  • Sgp ⁇ 2, Sgp syn , or Hgp syn was cotransfected with either of two SIV vectors, that still contained env, vif, vpr, vpx, tat, and rev in addition to the GFP reporter gene, but which contained large deletions in gag-pol.
  • the CEMxl74-SIN-SEAP cells which release SEAP after infection with SIN due to Tat transactivation (Means et al., 1997, J.Virol. 71, 7895- 7902), were infected with 1 ml of the supernatant of the transfected cells.
  • the titer of the RCR in the supernatant of cells transfected with Sgp ⁇ 2 and SIN-GFP ⁇ DP or Sgp ⁇ 2 and SIN-GFP ⁇ BP was determined to be 130 and 75 TCID 5 o/ml, respectively. This demonstrates that if RCR emerge at all with the synthetic gag-pol genes, the frequency is at least reduced by a factor of approximately 100 in comparison to a wild-type gag-pol expression plasmid.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Communicable Diseases (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Oncology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Saccharide Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The present invention relates to synthetic gag and gagpol genes optimized for high level expression via codon optimization and the uses thereof for the efficient generation of vector particles. The invention further relates to the generation of packaging cells and vaccines based on the synthetic gag and gagpol genes.

Description

Synthetic gagpol genes and their uses
The present invention relates to synthetic gag and gagpol genes optimized for high level expression via codon optimization and the uses thereof for the efficient generation of vector particles. The invention further relates to the generation of packaging cells and vaccines based on the synthetic gag and gagpol genes.
Retroviral based vector particles for gene therapy, retroviral based virus-like particles for vaccination or retroviral based DNA or RNA constructs for DNA vaccination should require the ability to express structural genes and enzymatic functions in high yields in order to obtain maximal efficiency and yet, at the same time, with lowest possible risk and side effects for the recipient. Efficient production of structural or enzymatic proteins from complex retroviruses like HTLN-1 and -2, Lentiviruses and Spumaviruses is limited by their complex regulatory mechanism of transcription, nuclear RΝA translocation and expression. For example the expression of SIN and HIN-1 late gene products is highly regulated both on transcnptional and post-transcriptional level therefore depending on the presence and function of early phase proteins like Tat and Rev. Whereas the anti-repressor function of Tat can be easily avoided by using heterologous transcnptional control via viral and/or cellular promotor/enhancer units, the expression of late genes, coding for structural or enzymatic functions (gag, env, pol) depends on the presence of the transactive Rev protein, known to promote the export of un- and partially spliced RΝAs from the nucleus to the cytoplasm via the interaction with its cognate RΝA recognition site Rev responsive-element (RRE), a complex of a 351 nucleotides (nt) long RΝA stem-loop structure located within the env open-reading-frame. Although Rev/RRE action is well accepted as a necessary prerequisite for HIN-1 late gene expression, the critical contribution of different cis-acting elements within the un- and singly spliced transcripts to Rev dependency and timely regulated expression is still controversially discussed. Accordingly, Rev dependent and timely regulated export of late singly or unspliced lentiviral mRΝAs has been explained in most reports either by inefficient splice site formation or attributed to inhibitory sequences located within the coding region (referred to as INS elements). Whereas the exact nature and function of so called INS elements is presently not completely clear, Rev dependent nuclear export and expression of the un- and singly spliced lentiviral mRNAs, at least when expressed from heterologous promotors, seems to engage and require the major splice donor site located in the untranslated region 5' of the Gag coding sequence (5'- UTR). This 5'-UTR also contains part of the RNA packaging signal (ψ), which - in contrast to the ψ-sites of traditional C and D-type retroviruses - also extends into the coding area of lentiviral genomes including the Gag and Pol reading frame.
Although lentiviral vector particles, virus-like particles and DNA or RNA constructs derived from complex retroviruses like HTLN-1 and -2, Lentiviruses and Spumaviruses offer great promise in the field of gene therapy, DΝA vaccination and/or vaccination, concerns regarding safety in humans still exist, due to the striking pathogenic potential of HTLN's and lentiviruses like HIN-1 or HIV-2. Moreover, due to the complex viral regulatory mechanisms underlying Gag and GagPol expression efficient expression is either limited by or depends heavily on the presence of cis-acting elements (UTR, RRE) and transactive proteins (Rev, Tat). Tat- independent transcription can be easily achieved by employing a constitutive mammalian promotor like the Cytomegalovirus immediate early promotor/enhancer unit. Rev independent expression of late HIN-1 genes such as those coding for structural or enzymatic proteins would be highly appreciable in order to improve efficiency and safety of lentiviral based gene therapy vector particles and DΝA vaccines as well as for antigen production in a mammalian expression system. Generally, two systems can be applied: (a) The Mason-Pfitzer monkey virus (MPMN) constitutive transport element (CTE), a cis-acting RΝA element located within the 3' untranslated region (UTR) of the viral genome, can substitute the HIN-1 Rev/RRE regulatory system. In addition, several other cis-acting RΝA elements of a variety of viruses also promoting nuclear export of intron containing RΝAs were discovered (e.g. Rous sarcoma virus, Simian retrovirus type D, Avian leukemia virus, HBN). Accordingly, the addition of the MPMV-CTE element or functional analogous cis-acting RΝA elements overcomes the effect of the so called INS elements, which have been proposed to account for the nuclear sequestration of late lentiviral RΝAs in the absence of Rev. (b) Low or no gene expression of late HIN-1 gene products in absence of Rev has been partially overcome by clustered single point mutations within the wobble positions of selected codons within coding DΝA sequences (Schwartz et άl. 1992a J. Virol. 66:7176-7182; Schneider et al. 1991, J. Nirol 71:4892-4903; Haas et al 1996, Curr. Biol.: 6:315-324). This approach aimed to destroy the so called INS elements in order to render the expression of the resulting Gag gene independent of Rev. However, the presence, nature and function of INS elements is still discussed controversially, indicating that a complete knockout of poorly characterized INS elements on the basis of single point mutations may be difficult to achieve. This view is confirmed by the observation of Qiu and colleagues (Qiu et al 1999 J Virol. Nov;73(l l):9145-52.), showing that the addition of a CTE element to the (only partially) altered Gag gene leads to a further increase of Gag expression. This phenomenon does not hold true for a completely Rev-independent Gag expression construct.
A preferred application of HTLN-1 and -2 as well as Lentivirus derived Gag and GagPol based expression constructs is DΝA vaccination and the production of antigens, preferably, virus-like particles, in higher eukaryotic expression systems like in mammalian and insect cells. Cell mediated immune responses to Gag and Pol products of HIV-1 were shown to protect from disease or at least to contribute to an efficient control of virus replication. Accordingly, in persons with chronic infection, HIV-1 -specific proliferative responses to p24 as well as the number of Gag specific CTL precursors were inversely related to the determined viral loads. Moreover, in CTL responses directed towards highly conserved epitopes within Gag or Pol seem to critically contribute to the control of virus replication in long-term non-progressing HIN infected individuals.
Different formats have been devised in the past towards presenting Gag and Pol containing immunogens to the immune system, recombinant virus-like particles (VLPs) and Gag/Pol containing DΝA or RΝA constructs being amongst them. VLPs were usually produced in a baculovirus driven insect cell expression system, which allows to bypass the complex regulation of Gag and Pol expression seen in the natural host or mammalian cells. Such VLPs turned out to be highly immunogenic in rodents as well as in nonhuman primates. However, to compile with the regulations of Good Manufacturing Practice (GMP) and in order to achieve proper posttranslational modification of Gag/Pol polypeptides (including coexpressed proteins that might be packaged in or presented by VLPs), expression of GagPol and derivatives thereof in mammalian cell lines would be highly appreciated.
Vaccination by direct injection of DΝA is a novel and promising method for delivering specific antigens for immunization. Plasmid DΝA immunization has potential advantages compared to traditional protein vaccination due to the strong T helper 1 (Thl) and CTL responses induced, the prolonged antigen expression and the long lived effector activity, and thus can be used for vaccination. In animal models, DNA vaccination has been shown to induce protective immunity against a variety of viral and parasitic pathogens. In most cases, strong, yet highly variable, antibody and cytotoxic T cell responses were associated with control of infection. Plasmid DNAs expressing genes derived from HIN-1 have been recently shown to induce humoral and cellular immune responses in rodents, in nonhuman primates and in phase I studies in humans. Although these constructs were able to induce an immune response, both circulating antibody titers and HIN-1 specific CTL levels were transient and low.
However, the development of DΝA or RΝA constructs and infectious, although not necessarily replication-competent, bacterial or viral vehicle encoding Gag or GagPol or derivatives for antigen production in eukaryotic cells and for vaccination purposes faces several limitations both, regarding safety and efficiency. Gag and GagPol expression using wildtype genes in the Rev-dependent situation is limited (a) by the complex viral regulatory mechanisms which involves several cis-acting elements (RRE, UTR) and trans-acting proteins (Rev, Tat). In absence of UTR, RRE, or Rev no or only minute amounts of Gag or GagPol protein will be produced. Gag or Gag/Pol expression therefore needs simultaneous expression of the Rev protein either from a bicistronic construct or from a separate plasmid. Both strategies limit the efficiency of generating cell lines in vitro or reduce the efficacy of DΝA vaccine constructs in vivo either due to an increase plasmid size or the necessity to transfect/transduce one single cell with both plasmids at the same time; and (b) by the presence of the Rev protein itself, acting as an RΝA shuttle between nucleus and therefore harboring an intrinsic risk. Moreover, when applied as a therapeutically vaccination in chronically HIV infected individuals, Rev could contribute towards reactivating latent viruses, thereby enhancing the infection, virus replication and disease.
Moreover, Gag and GagPol expression - irrespective of accomplished by the Rev/RRE system or mediated independent from Rev/RRE by addition of a constitutive transport element (CTE) or analogous cis acting sites - is limited by the presence of UTR, RRE and several stretches in the GagPol coding area, which are known to comprise the HIV-1 packaging signal ψ or other RΝA elements responsible for packaging the genomic RΝA into viral particles. These gag and gagpol nucleic acids comprising such RΝA elements could be packaged into and distributed by endogenous . retroviral particles and contribute towards the mobilization of endogenous retroviruses. Additionally, Gag or GagPol derived VLPs generated either in cell culture or in vivo following DNA vaccination would self package their own RNA, which precludes any safe application of such DNA vaccine constructs for vaccination purposes in humans.
Moreover, any kind of Gag and GagPol expression - irrespective of being achieved (i) by Rev/RRE system, (ii) by addition of CTE or analogous sites or (iii) after introduction of single point mutations in the Gag or GagPol gene - is limited by the wildtype sequence itself in the case of a Gag or GagPol DNA vaccine. Recombination events could occur between the vaccine construct itself (UTR, RRE, wildtype ORF) and endogenous retroviral sequences or, when administered as a therapeutic vaccine, between the nucleic acids of the vaccine construct and the genetic information of the virus circulating within the patient. Both could lead to the reconstitution of potentially infectious or chimeric virus particles inheriting unknown risks.
Another preferred application of lentiviral Gag and GagPol based vector particles is gene transfer into different types of dividing, resting or postmitotic cells. In contrast to standard retroviral gene transfer as mediated e.g. by Moloney Murine Leukemia Virus (MoMuLV) based vector particles, lentiviral gene transfer can be used for gene delivery into a variety of quiescent or non-dividing cells, such as neuronal, muscle, and liver as well as hematopoietic stem cells and might therefore considerably contribute to the prevention and treatment of genetic disorders, tumor diseases and infections. The transduction of non-dividing, resting or postmitotic cells as neuronal, hepatical tissue or hematopoietic progenitor cells by for the treatment of variety of genetic disorders or acquired diseases. The lentiviral vector particles are currently prepared by triple-transfection of a GagPol expression plasmid together with a transfer construct (containing the packaging signal and transfer gene; flanked by LTRs), and expression plasmid encoding an envelope protein derived from an amphotropic or xeno ropic retrovirus like Moloney Murine Leukemia Virus or Vesicular stomatitis virus into mammalian cell lines. Lentiviral vector particles from the supernatant of such transfected cells were able to stable transduce a large variety of different cells even after in vivo gene transfer. To increase the safety of these vector particles, most of the accessory genes of HIV-l were deleted from the packaging construct and the vector particles, thereby minimizing the risk for the emergence of potentially pathogenic replication competent recombinants.
However, the development of lentiviral Gag and GagPol based vector particles for gene transfer into quiescent or non-dividing cells faces several limitations regarding safety and efficiency. The currently used HIV-1 or SIV derived GagPol expression constructs contain parts of the 5'- untranslated region comprising the RNA packaging signal ψ. Although small deletions were introduced into the assumed packaging signal, it is unlikely that these deletions completely prevent packaging, since similar deletions in the context of an intact 5 '-leader sequence reduced packaging by less than a factor of 25 (Luban and Goff, 1994 J Virol. Jun;68(6):3784-93).
Moreover, if the GagPol expression construct can indeed be packaged by homologous or non- homologous recombination events between the viral RNA and the GagPol RNA of the expression construct during or after reverse transcription can not be excluded. This could lead to the - for safety reasons - undesired transfer of the GagPol gene (or parts of it) into target cells.
Moreover, there are two regions of homology between the GagPol expression constructs and the lentiviral transgene constructs: (1) Rev-dependent as well as CTE mediated, Rev independent GagPol expression (packaging proteins) requires either the complete UTR or at least part of it for efficient expression. Also the transgene constructs depend for efficient packaging on the RNA packaging signal that overlaps the 5' -UTR. (2) Since in HIN-1, SIV and other lentiviruses the packaging signal extends from the UTR into the gag gene (reviewed in Swanstrom-R and Wills- JW, 1997, Cold Spring Harbor: Cold Spring Harbor Laboratory Press, pp. 263-334.), the 5'- portion of the gag gene is part of many lentivirus derived transgene constructs. (3) Finally both, the transgene construct and Rev-dependent GagPol expression constructs contain the Rev- responsive element, since (i) RRE was suggested to include RΝA packaging functions and (ii) the transport of the transgene RΝA and the GagPol RΝA from the nucleus to the cytoplasm is Rev-dependent. These regions of extensive homology might facilitate homologous or non homologous recombination events.
The problem underlying the present invention is the development of retroviral based expression constructs allowing high level protein production in absence of transactive proteins and with lowest risk of self packaging of the construct or recombination events thereby avoiding the risk for reconstitution of potentially infectious or by other means hazardous particles. A further problem underlying the present invention is the provision of vaccines against diseases caused by Lentiviruses. The problem of the invention is solved by the subject matter defined in the claims.
The present invention is further illustrated by the figures.
FIG. 1:
Schematic representation of wildtype and synthetic gag encoding expression constructs. Open boxes indicate wildtype gag (wtgag) encoding genes including 3' and 5' located cis-acting sequences, whereas syngag without any 3' or 5' untranslated regions (UTR) is boxed black. Wtgag reading frames were fused to the cis-acting sequences 5' located UTR, the Rev responsive-element (RRE), the constitutive transport element (CTE), or combination thereof. To investigate the influence of the major splice donor (SD) on CTE mediated Rev-independent Gag expression, the UTR were mutated in order to destroy the splice-donor consensus sequence within this upstream located sequence, resulting in mutant ΔSD-wtgag-CTE.
FIG. 2:
Representation of syngag and wildtype gag open reading frames. (A) The Gag coding sequence was adapted to a codon usage occurring in highly expressed mammalian genes and aligned to the corresponding wildtype sequence shown above. Sequence identity between the synthetic and wildtype gene is indicated by lines. (B) The table indicates the different sequence composition of the wildtype and synthetic gag reading frame. The fully synthetic gag gene with an optimized codon usage shows a markedly increase in G/C content whereas the overall A/U content is reduced. Moreover codon usage adaptation increased the amount of immune-stimmulatory CpG islets (Pur-Pur-C-G-Pyr-Pyr) within the coding region from only 1 to 5.
FIG. 3:
HI 299 cells were transiently transfected with indicated constructs. For standardization Gag expression was compared to wildtype like Rev/RRE dependent expression mediated by cotransfection of a Rev expression construct (indicated as +). Rev-independent Gag expression was analysed by co-transfection of a negative control construct (vector; indicated as -). (A) Rev/RRE mediated wildtype expression. (B) CTE-mediated wildtype expression. (C) Synthetic expression using a codon optimized reading frame. Cells were harvested 48 hours post- transfection, lysed and 50 μg of total protein subjected to Western Blot analysis (A-C, lower panel). Yields of Pr55sag were measured by testing different dilutions of cell-lysate in a capture- ELISA using purified Pr55gag for standardization (A-C, upper panel) and were normalized by the
Pr55gas production obtained by autologous Rev/RRE dependent expression. Gag-expression levels obtained by transfection of different cell lines were performed to rule out cell type specific effects (D). Bars represent relative Pr55gag expression levels (taken UTR-wtgag-RRE+Rev as 100%) and are given as the mean of triplicate determinations.
FIG. 4:
Immunological analysis of an HLV-l Gag DNA vaccine based on synthetic and wildtype sequences. 80 μg of wildtype (wt-gag) and synthetic (syngag) encoding DNA expression constructs were injected intramuscularily (im) into BALB/c mice and boosted twice, after 3 weeks and 6 weeks and sacrificed after 7 weeks. (A) Strength of humoral responses induced in immunized BALB/c. Each bar represents the group mean (n=4) for anti-gag IgGl, anti-gag IgG2 and total IgG as determined by end-point dilution ELISA assay. End-point titers of the immune sera were defined as the reciprocal of the highest plasma dilution that resulted in an adsorbance value (OD 492) three times greater than that of a preimmune serum with a cut-off value of 0,05. (B) Cytotoxic T cell activity in splenocytes from mice immunized intramuscularly or subcutaneously by particle gun (p.g.) with gag expression constructs. Lymphoid cells obtained from mice five days after the boost injection were co-cultured with gag peptide-pulsed syngenic P815 mastocytoma cells (irradiated with 20,000 rad). Control assays included splenocytes of non immunized and wt-gag immunized mice stimulated in vitro with peptide-pulsed P815 cells. Cytotoxic effector populations were harvested after 5 days of in vitro culture. The cytotoxic response was read against gag-peptide pulsed A20 cells and untreated A20 negative target cells in a standard 51Cr release assay. Data shown were mean values of triplicate cultures.
FIG. 5:
Maps of the (B) SlV-derived packaging constructs based on optimized GagPol reading frames of SIN and HIV and (A) (A) and HIN-1 SlV-derived (B) transgene constructs or packaging constructs. Regions deleted from the SIN genome are marked by shaded boxes with the first and last deleted nucleotide given after the "Δ" sign (numbering according to Genbank entry M33262). Point mutations that inactivate reading frames are marked by asterisks, inactivated reading frames are marked in black. Synthetic GagPol reading frames are hatched. BGHpAd: bovine growth hormone polyadenylation signal; CMN: immediate early promoter/enhancer region from human cytomegalo virus; SFFN-Pr: Spleen focus forming virus U3 region; GFP: gene for the green fluorescence protein;
FIG. 6:
1-rnmunoblot analysis of cell culture supernatants harvested 3 days after transfection of HI 299 cells with the indicated lentiviral (HIN-1: Hgpsyn and SINmac23 : Sgpsyn) GagPol expression constructs and proviral DΝAs. Released GagPol particles were enriched by continuous sucrose density gradient sedimentation. Antigen peak fractions were separated on a denaturing 12,5% SDS-PAGE and analyzed by immunobloting using HIN-1 p24 (A) or SIV p27 specific (B) monoclonal antibodies. Protease activity and functionality in the polyprotein processing was shown by absence (-) or presence (+) of Saquinavir. Molecular weights of the detected precursor proteins (Pr) and mature cleavage products are indicated. (C) Additionally, antigen peak fractions were also tested for its content of HIV-l p24 capsid antigen using a commercially available capture ELISA format (DuPont, Boston, USA)
FIG. 7:
Transduction of growth-arrested cells. 293T cells were transfected with NiGΔBH, pHIT-G and the indicated lentiviral GagPol expression constructs. The MLN vector particle was generated by transfecting plasmids pLEGFP-Νl, pHIT60, and pHIT-G. Vector particle titers in the supernatant of transfected cells were determined on 293 cells in the presence of the indicated concentrations of aphidicolin. The titration was done in triplicates or quadruplicates. The means and the standard deviations are shown. GFU: green fluorescence forming units.
FIG. 8:
Detection of replication competent recombinants. CEMxl74-SIN-SEAP cells were infected with the supernatant of 293T cells co-transfected with SIN-GFPΔDP (A) or SIN-GFPΔBP (B) and the indicated gag-pol expression constructs. RLU: relative light units.
FIG. 9:
Schematical representation of a codon optimized GagPol derivative which can be used for DΝA vaccination. For safety and regulatory reasons the packaging signal Ψ was removed, the integrase deleted, and the reverse transciptase (RT) gene disrupted by insertion of a scrambled nef gene at the 3 'end of the DΝA sequence coding for the RT active site. The nef gene was dislocated by fusing its 5 'half to its 3 'half. Myristilation is of the GagPolΝef particles is inhibited by a Glycin to Alanin mutation at the myristilation site of the gag gene.
FIG. 10:
Positioning effect of codon optimized sequences on the level of Gag expression. (A) Representation of the Gag polyprotein (pi 7, p24, p7 and p6) and the composition of the codon optimized gene encoding Gag. Restriction sites that were introduced for convenient assembly of the codon optimised sequences are shown including their position relative to the ATG (A=l) start codon. The order of the cloned fragments is indicated as FI to F7 from the amino- to the carboxy-terminus of Gag. (B) Composition of chimeric Gag open reading frames. The nomenclature of the synthetic (syn-gag), the wild-type (wt-gag) and the chimeric gag genes (synl/wt2-7, synl-2/wt3-7, synl-3/wt4-7, syn4/wt5-7, wtl-4/syn5-7) are given on the left side. Used abbreviations reflect the composition of the gag genes, respectively, indicating fragments (1 to 7; (A)) that are based either on synthetic (syn-) or wild-type (wt-) sequences. A schematic representation of the gag gene variants is also given (central panel). Closed boxes indicate codon optimised fragments (syn-), open boxes wild-type (wt-) gag coding sequences. Intracellular Gag expression yields measured 2 days following transfection of H1299 cells with the gag gene variants cloned into a pCDNA3.1 vector are indicated at the right. Amounts of synthesized Gag protein were expressed as %p24 compared to p24 levels achieved after transfection of HI 299 cells with pCDNAsyn-gag.
The term "vector particle" as used herein refers to a transduction competent viral particle comprising packaging proteins, envelope proteins as exposed on the surface of the viral particle as well as an incorporated transfer construct.
The term "virus-like particle" as used herein refers to a transduction incompetent viral particle comprising at least packaging proteins such as gag or gagpol or derivatives thereof.
The term "construct" as used herein refers to an expression vector in the sense of molecular cloning like a plasmid, bacteriophage, phagemid, phasmids or the like.
The term "packaging construct" as used herein refers to constructs comprising the nucleic acid sequence necessary to produce packaging proteins in eukaryotic cells. The term "transfer construct" or "gene transfer construct" or "transgene construct" as used herein refers to constructs comprising the nucleic acid sequence necessary to produce an RNA containing the packaging site Ψ, the transfer gene and flanking LTRs or derivatives thereof. This RNA is capable to be incorporated within an transducing viral particle which itself is produced by an eukaryotic cell expressing packaging proteins. The LTRs and the packaging site Ψ is of retroviral, preferably, spumaviral or lentiviral and most preferably, of HIN-1, HIN-2, STV or FIV origin.
The term "transgene" as used herein refers to a nucleic acid which should be introduced in a cell as a therapeutic agent. This nucleic acid can serve e.g. as a decoy, triplehelix forming or antisense oligonucleotide, as a ribozyme, an aptamere, or as a nucleic acid inserted into the genom in a manner that ensures its function, replication and transmission as a normal gene.
The term "packaging proteins" as used herein refers to GagPol proteins of all retroviruses such as oncoviruses, HTLV-1 and 2, spumaviruses, lentiviruses, e.g. HIV-1, HIV-2, SIV, FIV, and in particular to proteins encoded by SEQ ID ΝO:2 (GagPol HlN-lπm) and SEQ ID ΝO:l (GagPol SlNm c239). which are capable of packaging an RΝA containing the packaging site Ψ such as an retroviral, preferably, lentiviral, most preferably, HIN-1, HIN-2, SIN, FIN genome and/or a transfer construct.
The term "HTV-IIΠB" refers to the HIN-1 isolate BH10 (Gen Bank Ace: M15654).
The term "synthetic gag or gagpol" as used herein refers to a complete or a partially codon usage optimized gene, whereas the partially codon optimized portion is located at the 5'-end. Furthermore, the codon optimized portion is strict or fully optimized in a sense that all codons were optimized according to table 1.
The present invention relates to nucleic acid sequences encoding the gag and pol polypeptides, whereby the amino acid Ala is encoded by the nucleic acid triplett gcc or get, Arg is encoded by agg or aga, Asn is encoded by aac or aat, Asp is encoded by gac or gat, Cys is encoded by tgc or tgt, Gin is encoded by cag or cat, Glu is encoded by gag or gaa, Gly is encoded by ggc or gga, His is encoded by cac or cat, He is encoded by ate or att, Leu is encoded by ctg or etc, Lys is encoded by aag or aat, Met is encoded by atg, Phe is encoded by ttc or ttt, Pro is encoded by ccc or cct, Ser is encoded by age or tec, Thr is encoded by ace or aca, Tip is encoded by tgg, Tyr is encoded by tac or tat, Val is encoded by gtg ot gtc and the stop codon is tga or taa.
Preferably, the present invention relates to nucleic acid sequences encoding the gag and pol polypeptides, whereby the amino acid Ala is encoded by the nucleic acid triplett gcc or get, Arg is encoded by agg or aga, Asn is encoded by aac or aat, Asp is encoded by gac or gat, Cys is encoded by tgc or tgt, Gin is encoded by cag or cat, Glu is encoded by gag or gaa, Gly is encoded by ggc or gga, His is encoded by cac or cat, He is encoded by ate or att, Leu is encoded by ctg or etc, Lys is encoded by aag or aat, Met is encoded by atg, Phe is encoded by ttc or ttt, Pro is encoded by ccc or cct, Ser is encoded by age or tec, Thr is encoded by ace or aca, Trp is encoded by tgg, Tyr is encoded by tac or tat, Val is encoded by gtg ot gtc and the stop codon is tga or taa, whereby the nucleic acid sequence in the region in which the reading frames encoding the gag and pol polypeptides overlap conesponds to the wildtype nucleic acid sequence encoding the gag and pol polypeptides.
Most preferably, the present invention relates to nucleic acid sequences as depicted in SEQ ID NO: l or 2.
We were able to demonstrate that consequent and strict codon optimization of the Gag and GagPol reading frames derived from lentiviruses, particularly SIV and HIV such as the Simian Immunodeficiency virus SlVmac 39 and the Human Immunodeficiency virus HIN-1 IΠB allowed high level expression of Gag, GagPol and derivatives thereof in complete absence of Rev/RRE, CTE and UTR which can be used directly for DΝA vaccination.
Moreover, we were able to demonstrate that consequent and strict codon optimization of lentiviruses, particularly SIV and HIV such as SιNmac 39 or HIN-1 IΠB GagPol reading frames with the exception where Gag and Pol reading frames are overlapping, allowed high level expression of GagPol and derivatives thereof in complete absence of Rev/RRE, CTE and UTR and can be used as packaging constructs for gene delivery.
In addition, we were able to demonstrate that strict and consequent codon usage optimisation of 5' sequences, but not 3' sequences of lentiviral gag genes, particularly those derived from SIN and HIN, e.g. SιNmac239 or H-N-liim, in particular from nucleotides 1-854, 1-697, 1-489, or 1-294 of the HIN-1 HXIO gag gene, are sufficient to direct Rev-independent Gag or GagPol expression.
Therefore, safe and efficient delivery and expression of Gag or GagPol genes according to the present invention allows (i) expression of high yields of native proteins in higher eukaryotes like in mammalian or insect cells and (ii) supporting the generation of transduction competent particles for vaccination and gene therapy purposes which is currently limited by the complex regulatory mechanisms of late Gag and GagPol expression.
The present invention is based on the observation, that the codon usage e.g. in the HIN genome (A/T content 55.88 %) significantly deviates from that of highly expressed mammalian genes (usually not exceeding 50%) indicating a regulatory function of the codon usage for timely regulated retroviral such as HTLN-1 and -2, onco viral, spumaviral and lentiviral gene expression.
Additionally, we were able to show that CTE-mediated, similar to the Rev/RRE mediated, expression of wildtype HIV-l late gene products (such as gag), essentially depends on the presence of the HIN-1 5 '-untranslated region (UTR) or at least parts of it, which also contains an RΝA stretch responsible for packaging of the genomic RΝA into the viral particle, and therefore touches sensible safety issues.
In order to eliminate inhibitory or otherwise regulatory sequences within retroviral such as such as HTLN-1 and -2, oncoviral, spumaviral and lentiviral Gag and GagPol genes, reading frames were optimized according to codon usage found in highly expressed mammalian genes. For that purpose, a matrix was generated considering almost exclusively those codons that are used most frequently and, less preferably, those that are used second most frequently in highly expressed mammalian genes as depicted in table 1. Using these codons from highly expressed human genes a fully synthetic reading frame not occurring in nature was created, which, however encodes the very same product as the original wildtype gene construct. Table I: Codon most frequently (codon 1) and second most frequently (codon 2) found in highly expressed mammalian genes.
Figure imgf000015_0001
(Ausubel et al 1994 Current Protocols in Molecular Biology 2, A1.8-A1.9).
Based on this matrix, the Wisconsin genetics computer gr-oup (gcg) software package was employed to create the synthetic reading frame by backtranslating the amino acid sequence of lentiviral Gag and GagPol polypeptides into the codon optimized synthetic reading frame. Few deviations from strict adherence to the usage of most frequently found codons may be made (i) to accommodate the introduction of unique restriction sites at approximately 250 to 300 nt intervals (see e.g. Fig. 2) (ii) to break G or C stretches extending more than 7 base pairs in order to allow consecutive PCR amplification and sequencing of the synthetic gene product. This may also be performed for the Gag and Pol based expression constructs used for DNA vaccination as well the packaging constructs used for gene transfer with exception of the latter where gag and pol reading frames are overlapping. In particular, it is preferred that the overlapping region of the gag and pol reading frames of the packaging constructs used for gene transfer may not be altered at position 1298 - 1558 of SEQ ID NO:2 and position 1175 - 1532 of SEQ ID NO:l, in order to maintain both reading frames.
In particular, the use of the synthetic Gag and Pol genes according to the invention comprising the GagPol genes of all Retroviruses such as Oncoviruses, Human T-cell Leukemia Virus 1 and 2, Spuma- and preferably, Lentiviruses, in particular derived from HIN and SIV as encoded by SEQ ID ΝO:2 (GagPol HIV-1IΠB) and SEQ ID NO:l (GagPol SINmac239) allows therefore the safe and efficient production of packaging proteins for delivering genetic information to cells, whereas codons of Gag and GagPol reading frames were adapted to highly expressed mammalian genes as specified above, except regions where gag and pol reading frames are overlapping.
In particular, the use of partially synthetic GagPol genes according to the invention comprising the GagPol genes of all retroviruses such as Oncoviruses, Human T-cell Leucemia Virus 1 and 2, Spuma- and preferably, lentiviruses, in particular HIV and SIV as encoded by chimeric Gag sequences as examplified in Fig. 10 allows therefore the safe and efficient production of packaging proteins for delivering genetic information into cells, whereas only an Ν-terminal portion of the GagPol genes were adapted to highly expressed mammalian genes.
The present invention allows therefore the safe and efficient production of packaging proteins by increasing the yields of GagPol synthesis as compared to the expression rates achieved from the wildtype gene or wildtype like genes characterized by clustered point mutations - if driven by the same cellular or viral promotor/enhancer unit - as a result of consequent adaptation of the codon usage to that of frequently expressed mammalian genes. The present invention allows the safe and efficient production of packaging proteins in complete absence of known cis-acting elements (UTR, RRE) or transactive proteins such as Rev and Tat.
The present invention further relates to a synthetic retroviral gag comprising a 5' sequence with strict and consequent codon usage optimisation and a remaining 3 '-sequence having the wildtyp sequence, the codon usage optimisation being sufficient to direct Rev-independent Gag or GagPol expression, wherein the codon optimized 5' region is at least from nucleotide 1 to 150, preferably from 1-294, preferably from 1-489, preferably from 1-697, or preferably from 1-854.
The present invention further relates to retroviral gag or gagpol based vector particles, whereby the packaging proteins are derived from retroviruses and are encoded by nucleic acids according to the invention and the transgene construct is derived from wildtype retroviruses.
The present invention further relates to retroviral gag or gagpol based vector particles, wherein the retrovirus is selected from the group of oncoviruses, HTLV-1 or -2, spumaviruses, lentiviruses, in particular HIV, in particular HIV-1, HlV-lms, HIV-2, and STV, in particular
SlNmac239-
The present invention further relates to retroviral gag or gagpol based vector particles, whereby the packaging proteins are derived from a first retrovirus and the transgene construct is derived from a different second retrovirus and whereby the packaging proteins are encoded by nucleic acids according to the invention.
The present invention further relates to retroviral gag or gagpol based vector particles, wherein the first and second retrovirus are selected from the group of oncoviruses, HTLN-1 and 2, spumaviruses, lentiviruses, in particular HIN, in particular HIN-1, HlV-lπm, HIV-2, and SIV, in particular SlNmac239.
The present invention further relates to retroviral gag or gagpol based vector particles generated by nucleic acid molecules comprising the nucleic acid sequence according to the invention having at least a 25 fold reduced incorporation rate of said nucleic acid molecules encoding the gag and pol polypeptides compared to retroviral particles generated by wildtype nucleic acid sequences encoding the gag and pol polypeptides. The present invention further relates to retroviral gag or gagpol based vector particles produced by nucleic acid molecules comprising the nucleic acid sequence according to the invention having at least a 100 fold reduced recombination rate between said nucleic acid molecules encoding the gag and pol polypeptides and a wildtype genome based transgene construct derived from the same or another retrovirus.
The present invention allows the efficient production of safe vector particles by reducing the opportunities for reconstituting infection competent hybrid retroviral particles by recombination events between nucleic acids encoding the packaging proteins and sequences encoding endogenous retroviral particles.
The present invention allows the efficient production of safe vector particles by reducing the risk for incorporating the RNA species encoding the packaging proteins into vector particles by more than 25 fold as compared to expression constructs containing the 5'-UTR or encoding the wildtype GagPol gene or by diminishing the risk of packaging the very same RNA into endogenous retroviral particles.
The present invention allows the efficient production of safe vector particles by reducing the risk of recombination between the packaging construct and the gene transfer construct by more than 100 fold, by combining a synthetic construct encoding the GagPol packaging function from one retrovirus e.g. HTLN-1 and -2, onco-, spuma- or lentivirus with the wildtype genome based transgene construct derived from (i) the very same or (ii) another retrovirus.
Thus, the present invention allows the efficient production of safe vector particles by combining the synthetic gene encoding the packaging proteins derived from a retrovirus, e.g. from lentiviruses, preferably from the HIV-1 or SIV, more preferably from the HrV-lmB or SINmac239, with the wildtype transgene construct based on the genome of the same retrovirus, e.g. from lentiviruses, preferably from the HIV-1 or SIV, more preferably from the H-N-lπm or SINmac239.
Furthermore, the present invention allows the efficient production of safe vector particles by combining the synthetic gene encoding the packaging proteins derived from a retrovirus, e.g. from lentiviruses, preferably from the HIN-1 or SIV, more preferably from the HIV-1 IΠB or SINmac239. with the wildtype transgene construct based on the genome of another retrovirus, e.g. from lentiviruses, preferably from the HIV-1 or SIV, more preferably from the HlN-luiB or SINmac239, thereby reducing homologies between transfer vector and packaging vector (as e.g. depicted in table 1 of example 4) resulting in safe and efficient packaging constructs being useful for the production of safe lentiviral vectors and gene delivery systems. Exemplary such vector particles may consist of a synthetic gene encoding the HIN-1 ΠIB derived packaging proteins with the wildtype SιNmac23 genome based transgene construct, or of a synthetic gene encoding the SIVmac239 derived packaging proteins with the wildtype HIV-1 ΠIB genome based transgene construct.
Synthetic GagPol genes may or may not encode the active integrase. In the latter case, the integrase gene may be deleted in total. Alternatively, the enzyme activity may by knocked out by one deletion or several short continuous in frame deletions of various length, preferably, not exceeding 5 amino acids and, more preferably, comprising a single codon. Also, the integrase may be rendered inactive by point mutations at various sites, preferably, by a single point mutation (e.g. position 3930 - 3932 in SEQ ID Νo:2 and position 3957 - 3959 in SEQ ID No:l are changed to glutamin encoding nucleotides).
The present invention furthermore relates to retroviral packaging cells for the generation of the vector particles according to the invention transformed with nucleic acid molecules comprising the nucleic acid sequence according to the invention.
GagPol packaging proteins according to the present invention may be stably, inducibly or transiently expressed in any primary or lab adapted cell or cell line of human, mammalian or non-mammalian origin. The synthetic GagPol genes may be episomal or chromosomal within these cells and may contain additional recombinant genes, preferable any specific fusogenic gene, more preferable any viral envelope gene, more preferable a retroviral envelope gene. These cells may also contain either an episomal or chromosomal transfer construct, but may not contain any additional viral wildtype sequences.
GagPol expression may be driven by any tissue- or cell type-specific promotor/enhancer such as muscle kreatin kinase or MHC class I promotor/enhancer or by any viral e.g. CMV immediate early, Rous Sarcoma Virus promotor/enhancer, Adenovirus major/late promotor/enhancer or nonviral e.g. synthetic promotor/enhancer.
GagPol expression may be either constitutive or inducible e.g. by addition of a specific inducer like ecdysone in the case of a hormone inducible promotor enhancer or by removal of a repressor protein (Tet on/off) or repressor gene (Cre/Lox).
GagPol based gene delivery into cells may be mediated by any means of transfecting or introducing a plasmid or plasmid derivative such as "midges" (closed linear DNA) encoding the synthetic GagPol gene or the above variants thereof into cells.
GagPol based gene delivery into cells may be also mediated by infectious recombinant (i) viral vectors such as recombinant Retroviruses, Vaccinia- or Poxviruses, Adenoviruses, Alphaviruses, Herpesviruses, Baculoviruses or any other recombinant virus, (ii) subviral components bridging transfection and infection procedures like e.g. Virosomes comprising nucleic acid/protein aggregates containing e.g. Influenca hemagglutinin, (iii) bacterial vectors such as recombinant Listeriae or Salmonellae or any other type of infectious, replicating or non-replicating vector.
Any means of introducing GagPol genes and derivatives thereof into cells may lead to a transient expression of GagPol. The introduction of the above indicated GagPol genes into cells may also allow the establishment of stable cell lines supporting either constitutive or inducible GagPol expression.
Retroviral, e.g. spumaviral and lentiviral, preferably HIN or SIN derived vector particles carrying the transgene RΝA and being capable of transducing nondividing, resting or postmitotic cells of various species in addition to dividing or proliferating cells may be generated by co- expressing a synthetic GagPol constructs according to the invention together with any appropriate amphotropic envelope like the vesticular stomatitis virus G protein, xenotropic envelope like the env protein of avian or murine sarcoma viruses e.g. the Rous sarcoma virus (RSV) Env protein, or cell type specific receptor protein like the HIV-1 Env protein, in the presence of any appropriate gene transfer construct encoding the desired packaging competent transgene RΝA. Transduction competent vector particles may be generated by co-transfecting or co-infecting or transfecting/infecting primary cells or cell lines of various origin such as mammalian Verocells,
HeLa, Cos, CHO or H1299 cells or insect SF9, High-5 or DS-2 cells with constructs encoding the synthetic GagPol derived packaging proteins, and any appropriate envelope or receptor structure as well as the desired transgene.
Alternatively, transduction competent vector particles may be generated by stable cell lines expressing GagPol from the synthetic GagPol gene according to the invention, either inducible or constitutively, whereby the stable cell lines may be co-transfected or co-infected or transfected/infected with constructs encoding any envelope or receptor structure as well as the desired transgene.
Preferably, transduction competent vector particles may be generated by stable cell lines expressing GagPol from the synthetic GagPol gene according to the invention together with any appropriate envelope or receptor structure, either inducible or constitutively, whereby the stable cell lines may be transfected or infected with constructs encoding the desired transgene.
Ideally, transduction competent vector particles may be obtained from a stable cell line mediating the expression of the synthetic GagPol gene and any appropriate receptor or envelope function together with the generation of the packaging competent transgene RNA, either in an inducible manner or constitutively or a combination thereof.
Most preferably, transduction competent vector particles are generated by transfection of cells with a plasmid or derivative thereof or by infecting cells with any infectious, although not necessarily replication competent, bacterial or viral vehicle encoding the synGagPol derived packaging proteins, and any appropriate envelope or receptor structure as well as the desired transgene.
Both transduction competent vector particles, as well as infectious, although not necessarily replication competent, bacterial or viral vehicle encoding the synGagPol derived packaging proteins, and any appropriate envelope or receptor structure as well as the desired transgene may be used for application ex vivo and in vivo. Furthermore, the present invention relates to nucleic acid molecules comprising the nucleic acid sequence according to the invention for use as an active pharmaceutical substance.
The present invention relates further to the use of nucleic acid molecules comprising the nucleic acid sequence according to the invention for the preparation of a pharmaceutical composition for the treatment and prophylactic of diseases caused by retroviruses.
Moreover, the present invention can be used for vaccination purposes, in particular, if used as nucleic acid based vaccination constructs, which have been shown to elicit a strong humoral and Th-1 like cellular immune response in mice. On the basis of the GagPol genes of the present invention it is now possible to construct effective retroviral, e.g. HTLV-1 and -2, lentiviral, e.g. comprising all known HIV and SIV clades and sequences as well as derivatives, preferably HIN- IIIIB or SrVmac239, based Gag and GagPol vaccines, thereby increasing the yields of GagPol production by the use of either fully or, as examplified in Fig. 10, partially codon optimized GagPol genes or derivatives thereof as compared to the expression rates achieved from the wildtype gene or wildtype like genes characterized by clustered point mutations - if driven by the same cellular or viral promotor/enhancer unit - as a result of consequent adaptation of the codon usage to that of frequently expressed mammalian genes
A preferred object of the present invention is the construction of retroviral GagPol based vaccines e.g. oncoviral, spumaviral or lentiviral, preferably HIN and SIN, more preferably HIN- IΠIB or SINmac23 based vaccines, especially modified also to improve safety of GagPol derived candidate vaccines if delivered e.g. as a DΝA or RΝA vaccine, by any kind of infectious, replicating or non replicating bacterial or viral vehicle or if administered as GagPol derived virus-like particles by consequent and strict codon usage adaptation in complete absence of known cis-acting elements or transactive proteins such as Rev and Tat.
Another preferred object of the present invention is the construction of retroviral GagPol based vaccines by reducing the opportunities for reconstituting infection competent hybrid retroviral particles by recombination events between nucleic acids encoding the packaging proteins and sequences encoding endogenous retroviruses. Another preferred object of the present invention is the construction of GagPol based vaccines by reducing the risk for incorporating the RNA species encoding Gag, GagPol or derivatives thereof into subviral Gag or GagPol derived virus-like particles by more than 25 fold as compared to expression constructs containing the 5' -UTR or encoding the wildtype Gag and GagPol derived genes and by diminishing the risk of packaging the very same RNA into endogenous retroviral particles thereby reducing the risk of recombination between the nucleic acid derived from a vaccine construct encoding Gag, GagPol or derivatives thereof according to the present invention with DNA or RNA derived from an infection by HIV, SIV or any lenti- or retrovirus or otherwise related virus.
This results in safe and efficient DNA or RNA constructs being useful for the production and delivery of safe GagPol derived vaccine constructs.
The present invention further relates to the nucleic acid sequence as depicted in SEQ ID NO:4.
The present invention further relates to nucleic acid molecules according to the invention, comprising an amino acid substitution wherein the myristilation of the gag-precursor is inhibited.
The present invention further relates to nucleic acid molecules according to the invention, wherein the HIV or SIV N-terminal amino acid glycin is substituted to aϊanin.
The present invention further relates to nucleic acid molecules according to the invention, wherein one nucleotide is added or two nucleotides are deleted to introduce a ribosomal frameshift so that the gag and pol coding regions are using the same reading frame.
The present invention further relates to nucleic acid molecules according to the invention, comprising a deletion of the complete protease gene or of a part of the protease gene or one or more point mutations in the protease gene so that the protease is rendered inactive.
The present invention further relates to nucleic acid molecules according to the invention, comprising a deletion of the complete reverse transcriptase gene or of a part of the reverse transcriptase gene or one or more point mutations in the reverse transcriptase gene so that the reverse transcriptase is rendered inactive. The present invention further relates to nucleic acid molecules according to the invention, comprising a deletion of the complete integrase gene or of a part of the integrase gene or one or more point mutations in the integrase gene so that the integrase is rendered inactive.
In order to further increase the safety of the vaccine constructs, codon optimized Gag or GagPol genes may be modified at distinct sites.
Preferably, the Gag or GagPol based synthetic gene is modified in such a way that the myristilation of Gag-precursors is inhibited (e.g. position 4 - 6 in SEQ ID No:2 and position 4 - 6 in SEQ ID No:l are changed to alanin or valin encoding nucleotides), thereby preventing budding of Gag or GagPol derived virus-like particles and increasing the amount of endogenously produced antigen.
Preferably, the Gag or GagPol based synthetic gene is modified in such a way that the Gag and Pol coding regions are using the same reading frame by introducing a ribosomal frameshift, e.g. by either introducing one nucleotide or deleting two nucleotides at any position, without creating a premature stop codon within the Gag coding region, preferably, between the position of the natural frameshift target site (slippery sequence; corresponding to position 1297 SEQ ID No:l and position 1294 SEQ TD No:2) and the Gag stop codon (specified by SEQ ID No:3), thereby increasing the amount of synthesized pol gene products.
Preferably, the Gag or GagPol based synthetic gene is modified in such a way that the protease (PR) activity is rendered inactive by a deletion comprising the complete or part of the PR gene or by one or several short continuous in frame deletions of various length, preferably, not exceeding 5 amino acids and more preferably, comprising a single codon, most preferably, encoding the aspartic acid at the PR active site. Also, the PR may be rendered inactive by point mutations at various sites, preferably, by a single point mutation any position, most preferably, at the active site (e.g. position 1632 - 1635 in SEQ ID No:2 and position 1575 - 1577 in SEQ JD No:l are changed to asparagin or alanin encoding nucleotides).
Preferably, the Gag or GagPol based synthetic gene is modified in such a way that the active site of the reverse transcriptase gene (RT) is rendered inactive by a deletion comprising the complete or part of the RT gene or one or several short continuous in frame deletions of various length, preferably, not exceeding 5 amino acids and more preferably, comprising a single codon encoding an amino acid at any position, most preferable within the active site being critical for RT function. Also, the RT may be rendered inactive by point mutations at various sites, preferably, by a single point mutation at the active site (e.g. position 2349 - 2351 in SEQ ID No:2 and position 2136 - 2138 in SEQ TD No:l are changed to asparagin or glutamic acid encoding nucleotides).
Preferably, the Gag or GagPol based synthetic gene is modified in such a way that the active site of the reverse transcriptase gene (RT) is dislocated e.g. at the very C-terminus of the synthetic GagPol gene in order to inhibit polymerase activity thereby preventing the production of potentially hazardous nucleotide sequences and/or reconstitution of infectious or by other means hazardous virus particles.
Preferably, the Gag or GagPol based synthetic reading frame is modified in such a way that the other lentiviral genes such as nef, rev, tat, which are rendered biological inactive (e.g. by gene scrambling), are inserted into selected parts of the Gag or GagPol reading frames, Preferably, into the active site of RT in order to broaden immunogenicity and safety.
Preferably, the Gag or GagPol based synthetic gene is modified in such a way that the potentially hazardous genes, like the integrase (IN) are deleted. Alternatively, the enzyme activity may by knocked out by one deletion or several short continuous in frame deletions of various length, preferably, not exceeding 5 amino acids and more preferably, comprising a single codon. Also, the integrase may be rendered inactive by point mutations at various sites, preferably, by a single point mutation (e.g. position 3930 - 3932 in SEQ ID No:2 and position 3957 - 3959 in SEQ TD No:l are changed to glutamin encoding nucleotides).
Alternatively, the Gag or GagPol based synthetic gene is modified by any combination of the above described possibilities. An example of a derivative of a HIN-lππ. GagPol based construct modified as described above, is depicted in Fig. 9 and specified in SEQ ID No: 4. The myristilation defective GagPol based derivative is at least as immunogen as the corresponding myristilation competent GagPol based derivative. Strict adherence to the codon usage of highly expressed mammalian genes also leads to an increase of the overall GC, GG and CC content and thereby introduces potentially immunomodulatory nucleic acid motifs that increase humoral and cellular immune responses at least 2-fold as compared to the corresponding wildtype gene or wildtype like genes that were rendered partially Rev independent by clustered point mutations.
The GagPol product or derivatives thereof encoded by the gagpol genes of the present invention may be stably, inducibly or transiently expressed in any primary or lab adapted cell or cell line of human, mammalian or non-mammalian origin in order to produce GagPol antigens, GagPol containing virus-like particles or derivatives thereof.
The expression of codon optimized GagPol or derivatives thereof in cell culture as well as in vivo following immunization with DNA or RNA constructs specified below may be driven by any tissue- or celltype-specific cellular promotor/enhancer such as muscle kreatin kinase or MHC class I promotor/enhancer or by any viral (e.g. CMN immediate early; Rous Sarcoma Virus promotor/enhancer, Adenovirus major/late promotor/enhancer) or nonviral e.g. synthetic promotor/enhancer.
GagPol expression may be either constitutive or inducible e.g. by addition of a specific inducer such as ecdysone in the case of a hormone inducible promotor enhancer or by removal of a repressor (Tet on/off) or repressor gene (Cre/Lox).
The delivery of codon optimized GagPol genes or derivatives thereof into cells in vitro may be mediated by any means of transfecting or introducing a construct, e.g. a plasmid or plasmid derivative such as "midges" (closed linear DΝA) encoding the synthetic GagPol gene or the above variants thereof into cells.
The delivery of codon optimized GagPol genes or derivatives in vivo may be mediated by injection of the construct, construct derivative or infectious, replicating or nonreplicating bacterial or viral vehicle into any site, preferably, rntramascularly, subcutaneously or intradermally, co-administered with any kind of adjuvant e.g. liposomes, ISCOMS, alum or via biodegradable particles, either directly or using technical devices like particle gun, biojector or by any other means. The delivery of codon optimized GagPol genes or derivatives into cells in vitro and in vivo may be also mediated by infectious or non infectious recombinant viral vehicles such as recombinant Retroviruses, Vaccinia- or Poxviruses, Adenoviruses, Alphaviruses, Herpesviruses, Baculoviruses or any other recombinant virus, subviral components bridging transfection and infection procedures like e.g. Virosomes comprising nucleic acid/protein aggregates containing e.g. Influenca hemagglutinin, or bacterial vectors such as recombinant Listeriae or Salmonellae or any other type of infectious, replicating or non-replicating vector.
Example 1:
Construction of the synthetic gag gene. All subsequent numbering of HIV-1 wildtype nucleotide sequences correspond to the HIV-1 isolate BH10 (GenBank Accession: M15654). All subsequent numbering of synthetic HIN-1 Gag encoding reading frames correspond to the start codon of the respective coding region. Position of restriction sites are defined by their cleavage site.
In order to eliminate inhibitory sequences within the HIN-1 gag reading frame, a synthetic gene was constructed, as described above, encoding the entire Pr55gag polyprotein via a codon-usage occurring most frequently in highly expressed mammalian genes. Few deviations from strict adherence to the optimized codon usage were made to accommodate the introduction of unique restriction sites at approximately 250 to 300 nt intervals (Seq ID No: 5). A comparison of synthetic and wildtype Gag coding sequences is shown in Fig. 2 A demonstrating that almost every wobble position within the wildtype coding region was altered by construction of the synthetic gene. The Wisconsin genetics computer group (gcg) software package was used to compare the varying composition of the wildtype and synthetic gag reading frame. Codon optimization resulted in an increased G/C and decreased A/U(T) content, as well as in a 5 fold increase in immune stimulatory CpG motifs (Fig. 2 B).
A synthetic sequence coding for the HIN-lπiB Pr55gag polyprotein was generated by translating the amino-acid sequence of the gag coding region (nucleotides 112 - 1650) into a synthetic-gag (syngag) coding sequence using a codon-usage occurring most frequently in mammalian cells as depicted in Seq ID No: 5. In order to fragmentize the syngag reading-frame, unique restriction sites were generated at positions 5 (NarT), 290 (StyT), 487 (Stul), 696 (SacE), 852 (EcoRV), 1107 (E-.tEII) and 1307 (BglT) by silent mutations. For cloning, additional restriction sites Kpnl (95), BamΗI, Xhol and Sαcl were introduced within non-coding regions. Synthetic fragments spanning regions between restriction sites KpnWIStyl (FI), StyVStul (F2), StuVEcόSN (F3), EcoRV/BstΕE (F4), BstEΩJBglil (F5) and Bgli/Sacl (F6) were created by stepwise PCR-amplification with overlapping 60 nt long oligonucleotides and subcloned using the pCR-Script™ Amp SK(+) Cloning Kit (Stratagene, Heidelberg, Germany) following the manufactures instructions. The unique restriction sites Kpnl, Sty , Stul, EcόKV, BstETl, Saul and S cl were used to assemble the fragments into a complete reading frame. Finally, full-length syngag was placed into the Kpnl and Xhol restriction sites of of pcDNA3.1(+) expression vector (Invitrogen, Leek, The Netherlands) under the transcriptional control of the immediate-early promoter-enhancer of the Cytomegalovirus (CMV) allowing constitutive transcription in mammalian cells.
Construction of chimeric Gag genes. Chimeric Gag genes were generated based on the synthetic HΓV-IΠIB derived synthetic gag-gene cloned into pCRscript Amp plasmid. Thus, different fragments of the synthetic gag-gene were replaced by wild type gag-sequences. For that purpose, wild type gag sequences corresponding to the codon optimised gag fragments that have been used to assemble the synthetic gag gene were amplified from an HX10 template using primer pairs that included restriction sites flanking the synthetic gene fragments, respectively (Fig. 10A, F1-F7). The chimeric Gag constructs shown in Fig. 10 were subcloned into a pcDNA 3.1+ expression vector.
Several wildtype expression vectors were established by means of standard molecular biology techniques in order to compare expression of synthetic and wildtype gag reading frames. Subgenomic HIN-1 wildtype sequences were cloned by PCR amplification or restriction digestion of HX10 provirus DΝA (Ratner et al. 1987, ATDS Res. Hum. Retroviruses. 1987. 3:57- 69). The wildtype gag sequence (nucleotides 9-1640) including a 5 '-located 103bp long UTR was cloned into the Kpnl aad hol restriction sites of pcDΝA3.1 (Stratagene) expression vector after PCR amplification of a 1667 nt fragment with primers utr-1 (5 '-gcg ggt ace gaa ttc cga cgc agg act egg ctt gc-3 ') and gag-2 (5 '-gcc gag etc etc gag gga tec tta ttg tga cga ggg gtc gtt gcc aaa gag-3') resulting in pCMV-UTR-wtgag. Wildtype gag was cloned into the Kpnl and Xhol restriction sites of pcDΝA3.1 (Stratagene) expression vector by PCR amplification of a 1537 nt fragment (103-1640) with primers gag-1 (5 '-gcg ggt ace gaa ttc agg aga gag atg ggt gcg aga gcg tea gta tta agc-3') and gag-2 resulting in pCMV-wtgag. PCR amplification of the UTR with primers utr-1 and utr-2 (5'-gga tgg cgc cca tct etc tec ttc tag cct cc-3') resulted in a 103 nt fragment (9-112) which was cloned into the Kpnl and Narl restriction sites of pCMN-syngag, thereby placing the UTR directly 5 ' of the start-codon of syngag resulting in pCMV-UTR- syngag. Bgϊ l and BamHl digestion of HX10 proviral DΝA released a 854 nt fragment (6976- 7830) containing the RRE and an inefficiently used splice acceptor site (pos. 7734). This RRE containing fragment was cloned into the BamΗϊ restriction site of the plasmids pCMV-UTR- wtgag, pCMV-wtgag, and pCMV-UTR-syngag resulting in pCMV-UTR-wtgag-RRE, pCMV- wtgag-RRE and pCMV-UTR-syngag-RRE, respectively. A DΝA fragment coding for the constitutive transport element (CTE) RΝA element of Simian Mason-Pfizer D-type retrovirus (MPMV, Accession Νr. M12349, nucleotides 7886 to: 8386), was amplified using MPMV proviral DΝA as template and primers cte-1 (5' get aGG ATC Ccc att ate ate gcc tgg aac 3') and cte-2 (5 ' cga aCT CGA Gca aac aga ggc caa gac ate 3 '). The 500 bp fragment was cloned into the -5αmHI and Xhol restrction sites of pCMV-wtgag and pCMV-UTR-wtgag resulting in constructs pCMN-wtgag-CTE and pCMV-UTR-wtgag-CTE respectively. A schematic representation of all constructs is summarised in Fig. 1 A.
Example 2:
Transfection of Gag encoding expression constructs into mammalian cells. Cells were transfected by the calcium coprecipitation technique (Graham and Van der EB 1973, Virol. 52, 456-467). The day prior to transfection 2xl06 cells were plated on 100 mm-diameter culture dishes and incubated for 24 hours. For transfection, 30μg of Pr55gag expressing plasmids was used and the total amount of DΝA adjusted to 45 μg with either pcDΝA3.1(+) vector or for co- transfection experiments with pCMV-rev. Cells were harvested 48 hours post transfection, washed two times in PBS and then further analyzed. Western blot analysis of cells transfected with Gag encoding expression constructs. Harvested cells were lysed in 0.5% Triton X-100, lOOmM Tris/HCl (pH7.4) subjected to repeated freeze/thaw cycles, cleared by centrifugation (20800 x g for 5 in.) and the total amount of protein measured by BIO-RAD Protein- Assay (Bio-Rad Laboratories, Munich, Germany) following the manufactures instructions. 50μg total protein were separated by electrophoresis on a denaturing SDS 12.5% polyacrylamide gel and transferred onto nitrocellulose membrane by electroblotting. Expression of Pr55gag was detected by a HIN-1 p24 specific monoclonal antibody 13-5 (Wolf et al. 1990, AEFO 1:24-29) and visualized by chromogenic staining. Gag capture-ELISA of cells transfected with Gag encoding expression constructs. The p24 capture-ELISA antibodies were kindly provided by Dr. Matthias Niedrig (RKI, Berlin, Germany). 96-well MaxiSorp ELISA plates (Nunc, Wiesbaden, Germany) were coated over night a with lOOμl of p24-specific monoclonal antibody ll-G-7 diluted 1:170 with 0.1 M carbonat-buffer (pH 9.5). The plates were washed 6 times with wash-buffer (0.1% Tween 20, 300 mM NaCl, 10 mM Na2HPO4 2H2O, 1.5 mM NaH2PO4 H2O). Different amounts of cell- lysats were diluted in 0.5% BSA in wash-buffer added to the wells and incubated over night at 4°C with a second horse-radish-peroxidase conjugated monoclonal antibody (diluted 1:600 in 0.5%> BSA in wash-buffer), recognizing a different epitope within p24. After washing the plates 6 times with wash-buffer, antibody conjugates were stained with OPD-solution (Abbott, Wiesbaden, Germany) and absorption OD (495 nm) measured with an ELISA Reader. The concentration of Pr55gag was determined by a calibration curve using different concentrations of purified Pr55gag, produced in insect cells using the baculovirus expression system (Wagner et al. 1994, Virology 200:162-175).
As expected, synthesis of Pr55ga from the wildtype gag gene (wtgag) was extremely low in absence of RRE and Rev (Fig. 3 A, lanes 5,6). However, following fusion of RRE downstream of wtgag gene and co-transfection of Rev, Pr55gas production increased only by factor of 1.5-2, suggesting that the Rev/RRE system per se was not sufficient to promote substantial Gag expression (Fig. 3 A, lanes 7, 8). In contrast, addition of the authentic 103 bp UTR 5' to the gag gene resulted in a 4-5 fold increase in protein production even in the absence of Rev (Fig. 3 A, lane 1, 3 and 4). Additionally, Rev/RRE interaction led to a further increase in Pr55gag production by a factor of 5-8 (2-4 ng/μg total protein; Fig. 3 A, lane 2) and by several orders of magnitude compared to wtgag-RRE co-transfected with Rev. Substitution of Rev/RRE by CTE- mediated nuclear export depended in very same way on the presence of the authentic UTR resulting on high level Gag expression.. However, CTE mediated Pr55gag production (Fig. 3 B, lane 3) reached only 70% - 80% ( 1,3 -1,6 ng/μg total protein) of the Rev-dependent Gag expression using Rev/RRE. (Fig. 3 B, lane 1), whereas in absence of UTR or only very little Gag was produced (Fig. 3 B, lanes 4, 5).
High level expression of Pr55gag was achieved in all cases after transient transfection of the syngag encoding plasmids (Fig. 3 C lane 1-4). Expression levels were not substantially altered by introducing the Rev/RRE system and was neither dependent nor influenced by the presence of UTR (Fig. 3 C lanes 3-4). Pr55gas expression levels exceeded those accomplished by CTE- mediated wtgag expression by more than 130%, and by about 50% to 100% by co-transfection of UTR-wtgag-RRE with Rev (Fig. 3 C, lane 6) and by orders of magnitude to those accomplished by wtgag-RRE whether or not Rev was co-transfected (Fig. 3 C, lane 7, 8). Therefore, codon usage adaptation allows Rev-independent expression in absence of any cis-acting regulatory elements resulting in high yields of Pr55gag production (3.5-6.5 ng/μg total protein).
These results were confirmed using various cell-lines (Cos7, HeLa) ruling out that cell-type specific factors critically contributed to the observed effects (Fig. 3 D).
Noteworthy, as indicated in Fig. 10, substitution of nucleotides 1-294 in the wild-type Gag gene by a strictly codon optimised synthetic gag sequence was sufficient to support substantial, Rev- independent Gag expression. This effect was even more pronounced, when the codon optimised fragment was extended to the 5' 489, 697 or 845 nucleotides. However, reversing the order of codon optimised and wild type sequences starting with 854 wild-type gag nucleotides followed by codon optimised gag-sequences resulted in a reduction of Gag expression to almost background levels. This strongly suggests that positioning strictly and consequently codon optimised gag sequences to the 5'- but not to the 3 '-end of the Gag gene clearly accounts for increased and Rev-independent Gag-expression.
Example 3:
Immunogenicity of Gag expression vectors after naked DNA vaccination. Female BALB/c mice (Charles River, Sulzfeld, Germany) were housed under specific pathogen-free conditions and injected at the age of 6 to 12 weeks. In order to evaluate the efficiency and type of immune response induced by either wildtype or synthetic reading frames after DNA vaccination, groups of each 3 mice received a subcutaneous (s.c.) primary injection, at the base of the tail, of a solution containing 100 μg plasmid DNA in a total volume of 100 μl PBS. At week 3 and 6 after the primary injection, mice received a boost injection, respectively.
Serum was recovered from mice by tail bleed at week two after the boost injection. Antibodies specific for the HIV Pr55gag polyprotein were quantified by an end-point dilution ELISA assay (in duplicate) on samples from individual animals. In brief, a solid phase of PrepCell purified Pr55g g proteins (100 μl of 1 μg/ml per well, overnight at 4° C in a refrigerator) was used to capture Pr55gag-reactive antibodies from the serum (2 h at 37°C), which were then detected with horseradish peroxidase (HRP)-conjugated goat anti-mouse IgGl, IgG2a and total IgG antibodies (1:2000 in PBS, 2% Tween 20, 3% FCS; 100 μl/well; PharMingen, Hamburg, Germany), followed by o-phenlyenediamine dihydrochloride solution (100 μl/well, 20 min at room temperature in the dark; Abbott Laboratories, Abbott Park, IL). End-point titers were defined at the highest serum dilution that resulted in an adsorbance value (OD 492) three times greater than that of the same dilution of a nonimmune serum. The serum of each mouse was assayed and these values were used to calculate the mean and standard deviation for each group of five mice. Single cell suspensions were aseptically prepared from spleens of mice 5 days after the boost immunization. Cells were suspended in α-MEM medium (Gibco) supplemented with 10 mM HEPES buffer, 5 x 10"5 β-mercaptoethanole and 10% FCS. 5% of a selected batch of Con A- stimulated rat spleen cell supernatants (Poggi et al. 1994, Eur J Immunol. 1994 Sep;24(9):2258- 61) were further added to the culture medium as a source of growth factors. Responder cells (3 x 107) were cocultured with 1.5 x 106 syngenic, V3LAI peptide-pulsed P815 cells (irradiated with 20,000 rad) in 10 ml tissue culture medium in upright 25 cm tissue culture flasks in a humidified atmosphere with 7% CO2 at 37°C. Cytotoxic effector populations were harvested after 6 days of in vitro culture. Serial dilutions of effector cells were cultured with 2 x 103 target cells in 200 μl round-bottom wells. Targets were autologous A20 cells (2 x 104/ml) incubated overnight at 37°C with 10"8 M of a 18-mer V3LAI peptide or a 23-mer P24CA AI peptide. Non peptide-pulsed cells were used as a negative control. Target and control A20 cells were labeled with 51Cr (1 hour at 37°C, 20 μCi/106 cells) prior to being added to the effector cells. After a 4 hour incubation at 37°C, 100 μl of supernatant were collected for γ-counting. The percentage specific release was calculated as [(experimental release-spontaneous release) / (total release- spontaneous release)] x 100. Total counts were measured after adding 1% Triton X-100 to the labeled target cells. Spontaneously released counts were always less than 20% of the total counts. Data shown are the mean of triplicate cultures. Standard errors of the mean of triplicate data were always less than 20% of the mean.
As depicted in Fig. 4, synthetic genes derived from lentiviral pathogens are capable of inducing a strong humoral and cellular immune response. Only in mice immunized with synthetic gag plasmids high levels of specific antibodies could be detected. A Th-1 response induces more likely a cellular immune response, which was verified by CTL chromium release assay of freshly prepared spleen cells of the immunized mice. Again, only mice immunized with a Gag expression plasmid encoded with an optimized codon usage was capable of inducing a strong cytolytic activity and therefore cell mediated immune response.
Example 4: Construction of synthetic genes coding for packaging functions of a lentiviral gene transfer vector system. All subsequent numbering of nucleotide sequences referring to synthetic GagPol sequences of HIV-1 or SIN correspond to the start codon of the respective coding region. Position of restriction sites are defined by their cleavage site.
Design of a codon optimized HIV-1 GagPol gene encoding packaging functions. A synthetic sequence coding for the HIV-1 ΠIB (BH10, GehBank Accession: M15654) Prl60GagPo1 polyprotein was generated by backtranslating the amino-acid sequence of the GagPol coding region (nucleotides 1 - 4343), except for the region where gag and pol reading frames are overlapping (nucleotides 1298 - 1539), and by using a codon-usage occurring most frequently in mammalian cells (as depicted in Seq ID Νo:2). In order to fragmentize this synthetic GagPol reading-frame (Hgpsyn), restriction sites were introduced at positions 290 (Styl), 487 (StuT), 1056 (PflMT), 1107 (BstETT), 1549 (Ball), 1852 (Saul), 2175 (Accl), 2479 (BstXT), 2682 (EcoNT), 2867 (SphT), 3191 (PfMT), 3370 (BclT), 3588 (Ball), 3791 (XmdTTT), and 4105 (Styl) by silent mutations. For cloning, additional restriction sites Kpnl, Xhol and Sαcl were introduced within non-coding regions. Synthetic fragments spanning regions between restriction sites Kpnl/Styl (FI), Styl/Stul (F2), StuVBsiEll (F3), BsfEΩJBall (F4), BaWSaul (F5), SauVAccl (F6), AccVBstXl (71), BstXl/EcoNl (F8), EcoWSphl (F9), SphVPfMl (F10), PfMTJBcH (Fll), BcWBaR (F12) BaWXmaUl (F13), XmaTTUStyl (F15) and StyVSacl (F16) were created by a stepwise PCR- amplification of overlapping 60 nt long oligonucleotides and subcloned using the pCR-Script™ Amp SK(+) Cloning Kit (Stratagene, Heidelberg, Germany) following the manufactures instructions. The unique restriction sites Kpnl, Styl, Stul, BsfEll, and Sαcl were used to assemble the fragments FI to F4 into one intermediate fragment (Fl-4). The unique restriction sites Kpnl, Ball, Saul and Sαcl were used to assemble the fragments F5 to F7 into a second intermediate fragment (F5-7). The unique restriction sites Kpnl, BstXT, EcoNl, Sphl, PflMT, BcR, Ball, XmdTTT, Styl and Sαcl were used to assemble the fragments F8 to F16 into a third intermediate fragment (F8-16). The intermediate fragments (Fl-4, F5-7 and pF8-16) were finally assembled into the full-length synthetic GagPol reading frame using restriction sites Kpnl/BstETT, BstElT/Accl, and AccVSacl respectively, and placed into the Kpnl and Sacl restriction sites of the pCR-Script™ Amp SK(+) cloning vector (Stratagene, Heidelberg, Germany). In order to obtain high level, constitutive expression in mammalian cells the synthetic GagPol coding region was placed into the Kpnl and Xhol restriction sites of pcDNA3.1(+) expression vector (Invitrogen, Leek, The Netherlands) under the transcriptional control of the immediate-early promoter- enhancer of the Cytomegalovirus (CMN) resulting in plasmid Hgpsyn (encoding HIN-1 GagPol). Design of a codon optimized SIV GagPol gene encoding packaging functions. A synthetic A synthetic sequence coding for the SrVmaC239 (SIVmm239, GenBank Accession: M33262) Prl60GagPo1 polyprotein was generated by backtranslating the amino-acid sequence of the GagPol coding region (nucleotides 1 - 4358), except the region where gag and pol reading frames are overlapping (nucleotides 1299 - 1533), and by using a codon-usage occurring most frequently in mammalian cells. In order to fragmentize this synthetic SIV derived GagPol reading frame, restriction sites were introduced at positions 120 (BstXT), 387 (XmaTTT), 650 (PstT), 888 (NdeT), 1224 (ApaT), 1422 (NspT), 1583 (EheT), 1899 (SauT), 2019 (BsaBT), 2372 (EcoNI), 2651 (SacTT), 2785 (BclT), 3025 (BamHT), 3268 (Styl), 3573 (BalT), 3745 (SauT), 3923 (PflMT), and 4188 (ApaT) by silent mutations. For cloning, additional restriction sites Kpnl, Xhol and Sαcl were introduced within non-coding regions. Synthetic fragments spanning regions between restriction sites KpnTI BstXT (Fl), BstXl/XmoTTl (F2), XmaTTVPstl (F3), PstVNdel (F4), NdeVApal (F5), ApάllNspl (F6), NspVEhel (TI), EheVSaul (F8), SauVBsaBl (F9), BsaBVEcoNl (F10), EcoNI/SαcII (Fll), SaclVBcR (F12), BcWBamHl (F13), BamHVStyl (F14), StyVBaR (F15), BaRJSaul (F16), SauVPflMl (Til), PflMUApal (F18) and ApaVSacl (F19) were created by a stepwise PCR-amplification of overlapping 60 nt long oligonucleotides and subcloned using the pCR-Script™ Amp SK(+) Cloning Kit (Stratagene, Heidelberg, Germany) following the manufactures instructions. The unique restriction sites Kpnl, BstXT, XmaTTl, Pstl, Ndel, Apa I and Sαcl were used to assemble the fragments fl to f6 into one intermediate fragments (fl-6). The fragments 17 to fl 1 were assembled into a second intermediate fragment (f7-l 1) using the unique restriction sites Kpnl, Ehel, Saul, BsaBl, EcoNI and Sαcl. The unique restriction sites Kpnl, BcR, BamHT, Styl, Ball, Saul, PflMT, Apal and Sαcl were used to assemble the fragments fl2 to fl9 into a third intermediate fragment (fl2-19). The intermediate fragments fl-6, f7-ll and fl2-19 were finally assembled into the full-length synthetic GagPol gene of SIN using restriction sites KpnVNspl, Nspl/SacU, and SacTl/Sacl respectively, and placed into the Kpnl and Sαcl restriction sites of the pCR-Script™ Amp SK(+) cloning vector (Stratagene, Heidelberg, Germany). In order to obtain high level, constitutive expression in mammalian cells the SIN derived synthetic GagPol gene was placed into the Kpnl and Xliol restriction sites of pcDΝA3.1(+) expression vector (Invitrogen, Leek, The Netherlands) under the transcriptional control of the immediate- early promoter-enhancer of the Cytomegalovirus (CMV) resulting in plasmid Sgpsyn (encoding SlVmac239 GagPol).
Codon usage of the 4.3 kb long GagPol genes of HIN-1 and SIN was optimized for expression in human cells by assembling the entire reading frame from synthetic oligonucleo tides. The synthetic genes encode GagPol proteins which have the same amino acid sequence as wildtype SIN or HIN-1 GagPol. On the nucleotide level there is an overall identity of 69,27 % for the synthetic and wildtype GagPol of HIN-1 and of 73,24% for the synthetic and wildtype SIN GagPol genes (Table 1).
Table 1: Genetic distances between the different SIV and HIV-1 GagPol coding sequences
Figure imgf000035_0001
Distance matrix of SrVmac239 and HINmB derived synthetic (syn) and wildtype (wt) GagPol coding sequences. Numbers indicate the percentage of missmatches between the compared GagPol sequences.
Optimizing the codon usage of HIN-1 GagPol reduces the homology to wildtype SIN GagPol by 41.48%) and vice versa (Table 1). The longest stretch of nucleotide identity between synthetic and wildtype GagPol is 357 bp in SIN and 306 bp in HIN-1. This stretch is located in the p6gag/p6po1 overlap region containing the slippery sequence and a stem loop structure, both of which are required for proper ribosomal frameshifting.
Example 5: Expression of synthetic GagPol genes. Rev-independent expression of the synthetic HIN-1 and SIN GagPol genes was assayed in transient transfection experiments followed by characterisation of the GagPol particles by conventional immunoblot analysis. Briefly, the day prior to transfection 2xl06 HI 299 cells were plated on 100 mm-diameter culture dishes and incubated for 24 hours. Cells were transfected using 45 μg of the synthetic GagPol expressing plasmids or proviral DNA (HX10 and SιNmaC239) with the calcium phosphate coprecipitation method as described (Graham and Nan der EB 1973, Nirol. 52, 456-467). Cell culture supernatants were harvested 72 hours post transfection. To block processing of GagPol, plated cells were cultured and transfected in the presence of 10 μM Saquinavir. GagPol particles were purified by separating the culture supernatants on 10-60% sucrose gradients as previously described (Wagner et al. 1994, Virology 200:162-175). GagPol particles banded at the expected density of 1.13 to 1.19 g/cm2, typical for HIV virions. The fractions with highest particle content were dialized against PBS and further separated by electrophoresis on a denaturing SDS 12.5% polyacrylamide gel prior to transfere onto nitrocellulose membrane by electroblotting. HIV-1 p24 capsid (CA) and SIN p27 CA and their precursor proteins were detected by the monoclonal antibodies 13-5 (Wolf et al. 1990, ATFO 1:24-29.) and 55-2F12 (NTH, Rockville, USA), respectively.
Western Blot analyses from supernatants of cells transfected with expression plasmids for the synthetic HIN-1 and SIN GagPol genes (Hgpsyn, Sgpsyn in Fig. 5 B) revealed efficient expression of GagPol in the absence of Rev (Fig. 5 A, B). The GagPol levels obtained with the synthetic genes were significantly higher as the levels obtained by transfection of infectious proviral constructs in the presence of Rev as assessed by a commercial capture ELISA format. Subviral particles were readily released from cells transfected with the synthetic GagPol expression plasmids and sedimentate at a density of about 1,16 g/ml, comparable to wildtype SIN or HIV-1 virions. No differences were observed in the processing of the GagPol precursor proteins encoded by the wildtype viruses and the corresponding synthetic GagPol genes (Fig. 5 A, B).
Example 6: Vector production and infection of target cells. The 293T cells were transfected with the calcium phosphate coprecipitation method as described (Graham and Van der EB 1973, Virol. 52, 456-
' 467). The plasmids pHIT60 (MLV GagPol expression plasmid), pHIT-G (VSV-G expression plasmid), SgpΔ2 (STV-GagPol expression plasmid also expressing vi vpr, vpx, tat and rev), and
ViGΔBH (self-inactivating SIV vector expressing the green fluorescent protein gene (GFP) under the control of the promoter and enhancer from spleen focus forming virus) have been described previously (Graham and Van der EB 1973, Virol. 52, 456-467; Fouchier et al. 1997, EMBO 16, 4531-4539). The Murine leukemia virus (MLV) vector pLEGFP-Nl, which contains the GFP gene under the control of an internal CMV promoter, was obtained from Clontech. The supernatant of the transfected cells was passed through a 0.45 μm filter and stored in aliquots at -80°C. The vector stocks were assayed for CA antigen levels using an HIV p24 enzyme-linked immunosorbent assay (ELISA) kit (Inno genetics, Gent, Belgium). The standard included in the ELISA kit was used for quantification of HIV-1 p24 CA antigen levels. Recombinant SIV p28 CA antigen from the SIN p28 antigen capture assay from the AIDS vaccine program (ΝCI- Frederic cancer research and development center) served as a standard for the quantification of SIN ρ28 levels.
To determine vector titers, 293 cells were seeded at 2.5 x 104 cells per well of a 24-well plate. One day later, the medium was removed and cells were incubated for 2 to 4 hours with serial dilutions of the vector preparations in a total volume of 200 μl. Fresh medium was added and the number of GFP positive cells was determined two days after infection. To arrest 293 cells in the Gl/S phase of the cell cycle, cells were seeded in a 96-well plate at a density of 2 to 5 x 104 cells per well at a concentration of 1, 3, or 5 μg aphidicolin/ml (Sigma). The titer of the vector stocks was determined on these cells as described above with aphidicohn being present during the entire culture period.
The functional integrity of the GagPol proteins being encoded by synthetic genes was analysed by cotransfection of the synthetic GagPol expression plasmids and an expression plasmid for NSN-G (pHIT-G) with the SIV vector ViGΔBH (Fig. 6). This is a self-inactivating vector, which expresses the green fluorescent protein gene (GFP) under the control of an internal promoter. The vector titers in the supernatant of the transfected cells were in the range of 1x10 GFP- forming units/ml supernatant (Table 2). Similar titers were obtained after cotransfection of ViGΔBH with a wildtype GagPol expression plasmid of SIV (SgpΔ2). The synthetic HIV-1 GagPol expression plasmid also allowed efficient transfer of the SIV vector (Table 2). The infectivity of the vector particles varied less than 2-fold between the different GagPol expression plasmids used (Table 2). Therefore, HIV-1 GagPol must recognise all czs-acting sequences of SIV required for packaging, reverse transcription, and integration with similar efficiency as SIV. As using packaging functions the results clearly extent a previous report on the packaging of HIV-2 RΝA, which is closely related to SlVmac, in HIN-1 particles (Kaye and Lever, 1998, J.Nirol. 72, 5877-5885). Table 2. Titer and infectivity of an SIV vector packaged by synthetic SIV and HIV-1 GagPol expression plasmids
Titera
Plasmids transfected Experiment 1 Experiment 2 Infectivityb
SgpΔ2, NiGΔBH, NSN-G 5.0xl06 1.4 x10° 5.9xl05
Sgpsyn, NiGΔBH, NSV-G l.OxlO6 2.7x105 3.4xl05
Hgpsyn, ViGΔBH, VSV-G 2.0xl06 2.0x10° 3.1xl05
ViGΔBH. VSV-G <5 <5 n.a. a GFP forming units (GFU) per ml supernatant of 293T cells transfected with the indicated plasmids; b GFU/ng CA antigen in the supernatant of transfected 293T cells; n.a.: not applicable.
Omitting the GagPol expression plasmid reduced the titer below the level of detection (Table 2), which argues against VSV-G mediated pseudotransduction. In addition, the percentage of GFP positive cells transduced with ViGΔBH, which had been packaged by Sgps n, Hgpsyn, or SgpΔ2, did not decrease significantly during a six week observation period (data not shown), which suggests integration of the vector into the host genome. As previously observed for another SIV vector packaged by SgpΔ2 and pHIT-G replication competent recombinants could not be detected during an 8 week observation period in ViGΔBH vector preparations packaged by SgpΔ2, Sgpsyn, or Hgpsyn and pHIT-G (data not shown). The transduction efficiency of vectors produced with the synthetic GagPol genes for non-dividing cells was assessed by arresting the target cells in the Gl phase of the cell cycle by aphidicolin treatment. The titer of an MLV-based vector in growth-arrested cells was reduced to background levels (Fig. 7). In contrast, the SIV vector titer was only slightly reduced by aphidicolin treatment. Νon-dividing cells could be transduced with the SIV vector independent of the GagPol expression plasmid used for the production of the vector particle (Fig. 7).
Example 7:
CEMxl74 cells (5 x 105) were infected for 4 hours with 0.5 ml of the vector preparation. The cells were washed three times with PBS and subsequently cultured in 5 ml medium. Infected cultures were split 1:5 to 1:10 twice weekly for eight weeks. The capsid antigen levels in theses cultures were determined 1 and 4 days, and 2, 4, 6 and 8 weeks after infection using the HIN- Innogenetics ELISA and the recombinant SIN p28 antigen as a standard. To determine the emergence of RCR on CEMxl74-SIN-SEAP cells (Means et al., 1997, J.Virol. 71, 7895-7902), lxlO5 cells were infected with 1 ml supernatant of the transfected cells for two hours and washed once in PBS. Infected cultures were cultured in 5 ml medium and split 1:5 to 1:10 twice weekly for four weeks. Secreted alkaline phosphatase activity in the supernatant of these cultures was determined at different time points after infection with the Phospha-Light-kit (Tropix, Bedford, MA, USA) as described by the manufacturer. The titer of RCRs was determined by limiting dilution. CEMxl74-STV-SEAP cells (2xl05 ) were incubated with 1 ml supernatant in a final volume of 2 ml. After two hours, six 200 μl aliquots were transferred to a 96-well plate and used as a starting point for 4-fold serial dilutions. Cultures were split 1:10 after one week. Syncytia formation was scored 1 and 2 weeks after infection and the TCID5o of the RCRs was calculated as described (Johnson and Byington, 1990, Quantitative assays for virus infectivity. In Techniques in HIN research. A. Aldovini and B.D. Walker, eds. (New York: Stockton Press), pp. 71-76. 1990).
Detection of replication-competent recombinants. The potential emergence of replication competent recombinants (RCR) from our vector preparation was analysed in CEMxl74 cells, which support replication of SIV and SIV hybrid viruses utilising a heterologous env gene (Reiprich et al, 1997, J.Virol. 71, 3328-3331). These cells were infected with 0.5 ml of NiGΔBH vector preparations (for titers see example 6, Table 2) packaged by SgpΔ2, Sgpsyn, or Hgps n and pHIT-G. Slightly elevated capsid antigen levels (<400 pg SINp27CA/ml) could be detected one and four days after infection. This has been observed previously and is probably due to carry over CA-antigen from the vector preparation (Schnell et al., 2000, Hum.Gene Ther. 11, 439- 447). More importantly, CA antigen levels were below the level of detection of 20 pg SJNp27/ml two to eight weeks after infection in all cultures and no cytopathic effects could be observed. Therefore, the titer of a potential RCR is below 2 infectious unit/ml. Since no RCR could be detected with the wild-type STV-gag-pol expression plasmid, SgpΔ2, this assay did not reveal potential advantages of the synthetic gag-pol expression plasmids. We therefore tested the frequency of emergence of RCR in an assay system, that would only require two homologous recombination events. SgpΔ2, Sgpsyn, or Hgpsyn was cotransfected with either of two SIV vectors, that still contained env, vif, vpr, vpx, tat, and rev in addition to the GFP reporter gene, but which contained large deletions in gag-pol. The CEMxl74-SIN-SEAP cells, which release SEAP after infection with SIN due to Tat transactivation (Means et al., 1997, J.Virol. 71, 7895- 7902), were infected with 1 ml of the supernatant of the transfected cells. Three days after infection elevated SEAP activity was observed in cultures infected with the SIN vectors packaged by the synthetic gag-pol expression plasmids (Fig. 8). This should be due to transcomplementation of gag-pol and subsequent transfer of the SIN vector. However, the SEAP activity returned to background levels during the next 3 weeks (Fig. 8) indicating the absence of RCR. In contrast, increasing SEAP activity three to ten days after infection revealed rapid emergence of RCR in SIN vectors packaged by SgpΔ2. This was accompanied by extensive syncytia formation and cell death. Using a limiting dilution approach, the titer of the RCR in the supernatant of cells transfected with SgpΔ2 and SIN-GFPΔDP or SgpΔ2 and SIN-GFPΔBP was determined to be 130 and 75 TCID5o/ml, respectively. This demonstrates that if RCR emerge at all with the synthetic gag-pol genes, the frequency is at least reduced by a factor of approximately 100 in comparison to a wild-type gag-pol expression plasmid.

Claims

Claims
1. Nucleic acid sequence encoding the gag and pol polypeptides, whereby the amino acid Ala is encoded by the nucleic acid triplet gcc or get, Arg is encoded by agg or aga, Asn is encoded by aac or aat, Asp is encoded by gac or gat, Cys is encoded by tgc or tgt, Gin is encoded by cag or cat, Glu is encoded by gag or gaa, Gly is encoded by ggc or gga, His is encoded by cac or cat, He is encoded by ate or att, Leu is encoded by ctg or etc, Lys is encoded by aag or aat, Met is encoded by atg, Phe is encoded by ttc or ttt, Pro is encoded by ccc or cct, Ser is encoded by age or tec, Thr is encoded by ace or aca, Trp is encoded by tgg, Tyr is encoded by tac or tat, Val is encoded by gtg ot gtc and the stop codon is tga or taa.
2. Nucleic acid sequence according to claim 1, whereby the nucleic acid sequence in the region in which the reading frames encoding the gag and pol polypeptides overlap corresponds to the wildtype nucleic acid sequence encoding the gag and pol polypeptides.
3. Nucleic acid sequence as depicted in SEQ TD NO:l or 2.
4. Nucleic acid sequence encoding the gag polypeptide, whereby the amino acid Ala is encoded by the nucleic acid triplet gcc or get, Arg is encoded by agg or aga, Asn is encoded by aac or aat, Asp is encoded by gac or gat, Cys is encoded by tgc or tgt, Gin is encoded by cag or cat, Glu is encoded by gag or gaa, Gly is encoded by ggc or gga, His is encoded by cac or cat, He is encoded by ate or att, Leu is encoded by ctg or etc, Lys is encoded by aag or aat, Met is encoded by atg, Phe is encoded by ttc or ttt, Pro is encoded by ccc or cct, Ser is encoded by age or tec, Thr is encoded by ace or aca, Trp is encoded by tgg, Tyr is encoded by tac or tat, Val is encoded by gtg ot gtc, and wherein said nucleic acid triplets are used only in the 5' sequence of the gag encoding nucleic acid, in particular from nucleotide 1 to 150, from 1-294, from 1-489, from 1-697, or from 1-854.
5. Retroviral gag or gagpol based vector particle, whereby the packaging proteins are derived from retroviruses and are encoded by nucleic acids according to claims 1 to 4 and the transgene construct is derived from wildtyp retroviruses.
6. Retroviral gag or gagpol based vector particle according to claim 5, wherein the retrovirus is selected from the group of oncoviruses, HTLV-1 or -2, spumaviruses, lentiviruses, in particular HIN, in particular HIN-1, HIN-lπiB, HIN-2, and SIV, in particular SINmac239.
7. Retroviral gag or gagpol based vector particle whereby the packaging proteins are derived from a first retrovirus and the transgene construct is derived from a different second retrovirus and whereby the packaging proteins are encoded by nucleic acids according to claims 1 to 4.
8. Retroviral gag or gagpol based vector particle according to claim 7, wherein the first and second retrovirus are selected from the group of oncoviruses, HTLN-1 and 2, spumaviruses, lentiviruses, in particular HJN, in particular HIN-1, HiN-lπm, HIN-2, and SIN, in particular SJNmaC239.
9. Retroviral gag or gagpol based vector particle generated by nucleic acid molecules comprising the nucleic acid sequence according to any one of claims 1 to 4 having at least a 25 fold reduced incorporation rate of said nucleic acid molecules encoding the gag and pol polypeptides compared to retroviral particles generated by wildtyp nucleic acid sequences encoding the gag and pol polypeptides.
10. Retroviral gag or gagpol based vector particle produced by nucleic acid molecules comprising the nucleic acid sequence according to any one of claims 1 to 4 having at least a 100 fold reduced recombination rate between said nucleic acid molecules encoding the gag and pol polypeptides and a wildtype genome based transgene construct derived from the same or another retrovirus.
11. Retroviral packaging cell for the generation of the retroviral vector particles according to any one of claims 5 to 10 transformed with nucleic acid molecules comprising the nucleic acid sequence according to any one of claims 1 to 4.
12. Nucleic acid sequence as depicted in -SEQ TD NO:4.
13. Nucleic acid molecule according to any one of claims 1 to 4 comprising an amino acid substitution wherein the myristilation of the gag-precursor is inhibited.
14. Nucleic acid molecule according to claim 13, wherein the HIV or SIN Ν-terminal amino acid glycin is substituted to alanin.
15. Nucleic acid molecule according to any one of claims 12 to 14, wherein one nucleotide is added or two nucleotides are deleted to introduce a ribosomal frameshift so that the gag and pol coding regions are using the same reading frame.
16. Nucleic acid molecule according to any one of claims 12 to 15 comprising a deletion of the complete protease gene or of a part of the protease gene or one or more point mutations in the protease gene so that the protease is rendered inactive.
17. Nucleic acid molecule according to any one of claims 12 to 16 comprising a deletion of the complete reverse transcriptase gene or of a part of the reverse transcriptase gene or one or more point mutations in the reverse transcriptase gene so that the reverse transcriptase is rendered inactive.
18. Nucleic acid molecule according to any one of claims 12 to 17 comprising a deletion of the complete integrase gene or of a part of the integrase gene or one or more point mutations in the integrase gene so that the integrase is rendered inactive.
19. Nucleic acid molecules comprising the nucleic acid sequence according to any one of claims 1 to 4 or 12 to 18 for use as an active pharmaceutical substance.
20. Use of nucleic acid molecules comprising the nucleic acid sequence according to any one of claims 1 to 4 or 12 to 18 for the preparation of a pharmaceutical composition for the treatment and prophylactic of diseases caused by retroviruses.
PCT/EP2001/005744 2000-05-18 2001-05-18 Synthetic gagpol genes and their uses WO2001088141A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
DE60121572T DE60121572T2 (en) 2000-05-18 2001-05-18 SYNTHETIC GENES FOR GAGPOL AND ITS USES
AU2001281781A AU2001281781A1 (en) 2000-05-18 2001-05-18 Synthetic gagpol genes and their uses
US10/276,482 US7378515B2 (en) 2000-05-18 2001-05-18 Synthetic gagpol genes and their uses
BR0110706-2A BR0110706A (en) 2000-05-18 2001-05-18 Synthetic gagpol genes and their uses
EP01960229A EP1282712B1 (en) 2000-05-18 2001-05-18 Synthetic gagpol genes and their uses
CA002408433A CA2408433A1 (en) 2000-05-18 2001-05-18 Synthetic gagpol genes and their uses
HK03105174A HK1053853A1 (en) 2000-05-18 2003-07-17 Sythetic gagpol genes and their uses
US11/754,755 US8287881B2 (en) 2000-05-18 2007-05-29 Synthetic gagpol genes and their uses
US13/615,587 US20130122585A1 (en) 2000-05-18 2012-09-14 Synthetic gagpol genes and their uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP00110623.6 2000-05-18
EP00110623A EP1156112B1 (en) 2000-05-18 2000-05-18 Synthetic gagpol genes and their uses

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10276482 A-371-Of-International 2001-05-18
US11/754,755 Continuation US8287881B2 (en) 2000-05-18 2007-05-29 Synthetic gagpol genes and their uses

Publications (2)

Publication Number Publication Date
WO2001088141A2 true WO2001088141A2 (en) 2001-11-22
WO2001088141A3 WO2001088141A3 (en) 2002-04-18

Family

ID=8168767

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2001/005744 WO2001088141A2 (en) 2000-05-18 2001-05-18 Synthetic gagpol genes and their uses

Country Status (10)

Country Link
US (3) US7378515B2 (en)
EP (2) EP1156112B1 (en)
CN (1) CN1429270A (en)
AT (2) ATE318908T1 (en)
AU (1) AU2001281781A1 (en)
BR (1) BR0110706A (en)
CA (1) CA2408433A1 (en)
DE (2) DE60026199T2 (en)
HK (2) HK1042319B (en)
WO (1) WO2001088141A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003025003A2 (en) * 2001-09-20 2003-03-27 Glaxo Group Limited Hiv-gag codon-optimised dna vaccines
EP1729801A2 (en) * 2004-03-31 2006-12-13 Advanced Bioscience Laboratories, Inc. Mutant viral nucleic acids and vaccine containing same
WO2007012691A1 (en) * 2005-07-27 2007-02-01 Consejo Superior De Investigaciones Científicas Recombinant vectors based on the modified ankara virus (mav) as preventive and therapeutic vaccines against aids
WO2013156443A1 (en) 2012-04-17 2013-10-24 F. Hoffmann-La Roche Ag Method for the expression of polypeptides using modified nucleic acids
US9988425B2 (en) 2012-01-27 2018-06-05 Laboratories Del Dr. Esteve S.A. Immunogens for HIV vaccination
WO2020260431A1 (en) 2019-06-28 2020-12-30 F. Hoffmann-La Roche Ag Method for the production of an antibody
US11666651B2 (en) 2019-11-14 2023-06-06 Aelix Therapeutics, S.L. Prime/boost immunization regimen against HIV-1 utilizing a multiepitope T cell immunogen comprising Gag, Pol, Vif, and Nef epitopes

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002022080A2 (en) * 2000-09-15 2002-03-21 Merck & Co., Inc. Enhanced first generation adenovirus vaccines expressing codon optimized hiv1-gag, pol, nef and modifications
EP1201750A1 (en) * 2000-10-26 2002-05-02 Genopoietic Synthetic viruses and uses thereof
US7410795B2 (en) 2002-12-23 2008-08-12 Vical Incorporated Codon-optimized polynucleotide-based vaccines against human cytomegalovirus infection
DE10260805A1 (en) * 2002-12-23 2004-07-22 Geneart Gmbh Method and device for optimizing a nucleotide sequence for expression of a protein
JP4550115B2 (en) 2004-08-03 2010-09-22 ジーンアート・アクチエンゲゼルシャフト Method for regulating gene expression by changing CpG content
DE102004037611B4 (en) * 2004-08-03 2013-10-02 Geneart Ag Inducible gene expression
US8323891B2 (en) * 2008-08-01 2012-12-04 Claflin University miRNA triplex formations for the downregulation of viral replication
CA2793959C (en) 2010-03-25 2019-06-04 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
HUE037408T2 (en) 2011-06-10 2018-08-28 Univ Oregon Health & Science Cmv glycoproteins and recombinant vectors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998034640A2 (en) * 1997-02-07 1998-08-13 Merck & Co., Inc. Synthetic hiv gag genes
WO2000015819A1 (en) * 1998-09-11 2000-03-23 The Children's Medical Center Corporation Packaging cell lines for hiv-derived retroviral vector particles
WO2000039304A2 (en) * 1998-12-31 2000-07-06 Chiron Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998034640A2 (en) * 1997-02-07 1998-08-13 Merck & Co., Inc. Synthetic hiv gag genes
WO2000015819A1 (en) * 1998-09-11 2000-03-23 The Children's Medical Center Corporation Packaging cell lines for hiv-derived retroviral vector particles
WO2000039304A2 (en) * 1998-12-31 2000-07-06 Chiron Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
GRAF M ET AL: "Concerted action of multiple cis-acting sequences is required for rev dependence of late human immunodeficiency type 1 gene expression" JOURNAL OF VIROLOGY, vol. 74, no. 22, November 2000 (2000-11), pages 10822-10826, XP002182672 AMERICAN SOCIETY FOR MICROBIOLOGY US *
KOTSOPOULOU E ET AL: "A Rev-independent human immunodeficiency virus type 1 (HIV-1)-based vector that exploits a codon-optimized HIV-1 gag-pol gene" JOURNAL OF VIROLOGY, vol. 74, no. 10, May 2000 (2000-05), pages 4839-4852, XP002152133 AMERICAN SOCIETY FOR MICROBIOLOGY US *
See also references of EP1282712A2 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003025003A2 (en) * 2001-09-20 2003-03-27 Glaxo Group Limited Hiv-gag codon-optimised dna vaccines
WO2003025003A3 (en) * 2001-09-20 2003-12-04 Glaxo Group Ltd Hiv-gag codon-optimised dna vaccines
JP2005511019A (en) * 2001-09-20 2005-04-28 グラクソ グループ リミテッド HIV-GAG codon optimized DNA vaccine
EP2322626A1 (en) * 2001-09-20 2011-05-18 Glaxo Group Limited HIV-GAG codon-optimised DNA vaccines
EP1729801A4 (en) * 2004-03-31 2010-01-13 Advanced Bioscience Lab Inc Mutant viral nucleic acids and vaccine containing same
EP1729801A2 (en) * 2004-03-31 2006-12-13 Advanced Bioscience Laboratories, Inc. Mutant viral nucleic acids and vaccine containing same
ES2281252A1 (en) * 2005-07-27 2007-09-16 Consejo Superior De Investigaciones Cientificas Recombinant vectors based on the modified ankara virus (mva) as preventive and therapeutic vaccines against aids
WO2007012691A1 (en) * 2005-07-27 2007-02-01 Consejo Superior De Investigaciones Científicas Recombinant vectors based on the modified ankara virus (mav) as preventive and therapeutic vaccines against aids
US9988425B2 (en) 2012-01-27 2018-06-05 Laboratories Del Dr. Esteve S.A. Immunogens for HIV vaccination
US10815278B2 (en) 2012-01-27 2020-10-27 Laboratorios Del Dr. Esteve S.A. Immunogens for HIV vaccination
US11325946B2 (en) 2012-01-27 2022-05-10 Laboratorios Del Dr. Esteve S.A. Method of treating HIV-1 infection utilizing a multiepitope T cell immunogen comprising gag, pol, vif and nef epitopes
US11919926B2 (en) 2012-01-27 2024-03-05 Esteve Pharmaceuticals, S.A. Method of treating HIV-1 infection utilizing a multiepitope T cell immunogen comprising Gag, Pol, Vif, and Nef epitopes
WO2013156443A1 (en) 2012-04-17 2013-10-24 F. Hoffmann-La Roche Ag Method for the expression of polypeptides using modified nucleic acids
WO2020260431A1 (en) 2019-06-28 2020-12-30 F. Hoffmann-La Roche Ag Method for the production of an antibody
US11666651B2 (en) 2019-11-14 2023-06-06 Aelix Therapeutics, S.L. Prime/boost immunization regimen against HIV-1 utilizing a multiepitope T cell immunogen comprising Gag, Pol, Vif, and Nef epitopes

Also Published As

Publication number Publication date
EP1156112A1 (en) 2001-11-21
US8287881B2 (en) 2012-10-16
AU2001281781A1 (en) 2001-11-26
EP1156112B1 (en) 2006-03-01
BR0110706A (en) 2003-01-28
ATE318908T1 (en) 2006-03-15
US20130122585A1 (en) 2013-05-16
DE60026199D1 (en) 2006-04-27
CA2408433A1 (en) 2001-11-22
US20070293448A1 (en) 2007-12-20
WO2001088141A3 (en) 2002-04-18
US7378515B2 (en) 2008-05-27
US20040152069A1 (en) 2004-08-05
HK1042319B (en) 2006-10-20
ATE333503T1 (en) 2006-08-15
DE60121572D1 (en) 2006-08-31
HK1053853A1 (en) 2003-11-07
EP1282712B1 (en) 2006-07-19
CN1429270A (en) 2003-07-09
DE60121572T2 (en) 2007-06-28
HK1042319A1 (en) 2002-08-09
EP1282712A2 (en) 2003-02-12
DE60026199T2 (en) 2006-11-23

Similar Documents

Publication Publication Date Title
US8287881B2 (en) Synthetic gagpol genes and their uses
US7981671B2 (en) Use of triplex structure DNA in transferring nucleotide sequences
JP5033303B2 (en) Polynucleotides encoding polypeptides with antigenic type C HIV, polypeptides and uses thereof
Guy et al. Mutational analysis of the HIV nef protein
JP5178196B2 (en) Recombinant MVA virus expressing the clade A / G, clade B, and clade C modified HIVenv, gag, and pol genes
JP2012147792A (en) Polynucleotide encoding antigenic hiv type c polypeptide, polypeptide and use thereof
JP2009082136A (en) Polynucleotide encoding antigenic hiv type b polypeptide and/or antigenic hiv type c polypeptide, and polypeptide and use thereof
CA2439067C (en) Lentiviral packaging constructs
KR19990087126A (en) Synthetic Human Immunodeficiency Virus Gene
AU3339195A (en) Protein targeting into hiv virions based on hiv-1 vpr fusion molecules
US20090227658A1 (en) Methods and compositions for immunization against hiv
JP2005521380A (en) Polynucleotide encoding antigenic type B HIV polypeptide, polypeptide and use thereof
JP4627950B2 (en) Molecular clone with mutant HIV gag / pol, SIV gag and SIV env genes
JPH01500962A (en) A viral vector, a DNA recombinant encoding the F protein of the AIDS-causing virus, a cell culture infected with the vector, a method for producing the above protein, the obtained protein, a vaccine, and the obtained antibody
JP5324912B2 (en) HIVVif mutant
EP1543837A1 (en) Virus-like particle (VLP) as vaccine
Vogt et al. Heterologous HIV-2 challenge of rhesus monkeys immunized with recombinant vaccinia viruses and purified recombinant HIV-2 proteins
CA2372456C (en) Viral infection inhibitor targeting integrase n-terminal domain
Young Construction, characterization and immunogenicity of human immunodeficiency virus-like particles
AU5543700A (en) Siv-based packaging-defficient vectors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2408433

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 01809614X

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2001960229

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001960229

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10276482

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: JP

WWG Wipo information: grant in national office

Ref document number: 2001960229

Country of ref document: EP